Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Pharmacol ; 103(3): 158-165, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36460345

RESUMEN

Cisplatin is an effective chemotherapeutic agent, yet its use is limited by several adverse drug reactions, known as cisplatin-induced toxicities (CITs). We recently demonstrated that cisplatin could elicit proinflammatory responses associated with CITs through Toll-like receptor 4 (TLR4). TLR4 is best recognized for binding bacterial lipopolysaccharide (LPS) via its coreceptor, MD-2. TLR4 is also proposed to directly bind transition metals, such as nickel. Little is known about the nature of the cisplatin-TLR4 interaction. Here, we show that soluble TLR4 was capable of blocking cisplatin-induced, but not LPS-induced, TLR4 activation. Cisplatin and nickel, but not LPS, were able to directly bind soluble TLR4 in a microscale thermophoresis binding assay. Interestingly, TLR4 histidine variants that abolish nickel binding reduced, but did not eliminate, cisplatin-induced TLR4 activation. This was corroborated by binding data that showed cisplatin, but not nickel, could directly bind mouse TLR4 that lacks these histidine residues. Altogether, our findings suggest that TLR4 can directly bind cisplatin in a manner that is enhanced by, but not dependent on, histidine residues that facilitate binding to transition metals. SIGNIFICANCE STATEMENT: This work describes how the xenobiotic cisplatin interacts with Toll-like receptor 4 (TLR4) to initiate proinflammatory signaling that underlies cisplatin toxicities, which are severe adverse outcomes in cisplatin treatment. Here, this study provides a mechanistic bridge between cisplatin extracellular interactions with TLR4 and previous observations that genetic and chemical inhibition of TLR4 mitigates cisplatin-induced toxicity.


Asunto(s)
Cisplatino , Receptor Toll-Like 4 , Animales , Ratones , Alérgenos , Cisplatino/toxicidad , Histidina , Lipopolisacáridos/farmacología , Antígeno 96 de los Linfocitos/química , Antígeno 96 de los Linfocitos/genética , Antígeno 96 de los Linfocitos/metabolismo , Receptor Toll-Like 4/efectos de los fármacos , Receptor Toll-Like 4/metabolismo
2.
EMBO Rep ; 22(5): e51280, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33733573

RESUMEN

Toll-like receptor 4 (TLR4) recognizes bacterial lipopolysaccharide (LPS) and can also be activated by some Group 9/10 transition metals, which is believed to mediate immune hypersensitivity reactions. In this work, we test whether TLR4 can be activated by the Group 10 metal platinum and the platinum-based chemotherapeutic cisplatin. Cisplatin is invaluable in childhood cancer treatment but its use is limited due to a permanent hearing loss (cisplatin-induced ototoxicity, CIO) adverse effect. We demonstrate that platinum and cisplatin activate pathways downstream of TLR4 to a similar extent as the known TLR4 agonists LPS and nickel. We further show that TLR4 is required for cisplatin-induced inflammatory, oxidative, and cell death responses in hair cells in vitro and for hair cell damage in vivo. Finally, we identify a TLR4 small molecule inhibitor able to curtail cisplatin toxicity in vitro. Thus, our findings indicate that TLR4 is a promising therapeutic target to mitigate CIO.


Asunto(s)
Antineoplásicos , Neoplasias , Ototoxicidad , Antineoplásicos/efectos adversos , Cisplatino/toxicidad , Humanos , Neoplasias/tratamiento farmacológico , Platino (Metal)/uso terapéutico , Receptor Toll-Like 4/genética
3.
Int J Mol Sci ; 23(13)2022 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-35806229

RESUMEN

Cisplatin is a platinum-based chemotherapeutic that has long since been effective against a variety of solid-cancers, substantially improving the five-year survival rates for cancer patients. Its use has also historically been limited by its adverse drug reactions, or cisplatin-induced toxicities (CITs). Of these reactions, cisplatin-induced nephrotoxicity (CIN), cisplatin-induced peripheral neuropathy (CIPN), and cisplatin-induced ototoxicity (CIO) are the three most common of several CITs recognised thus far. While the anti-cancer activity of cisplatin is well understood, the mechanisms driving its toxicities have only begun to be defined. Most of the literature pertains to damage caused by oxidative stress that occurs downstream of cisplatin treatment, but recent evidence suggests that the instigator of CIT development is inflammation. Cisplatin has been shown to induce pro-inflammatory signalling in CIN, CIPN, and CIO, all of which are associated with persisting markers of inflammation, particularly from the innate immune system. This review covered the hallmarks of inflammation common and distinct between different CITs, the role of innate immune components in development of CITs, as well as current treatments targeting pro-inflammatory signalling pathways to conserve the use of cisplatin in chemotherapy and improve long-term health outcomes of cancer patients.


Asunto(s)
Antineoplásicos , Neoplasias , Enfermedades del Sistema Nervioso Periférico , Antineoplásicos/efectos adversos , Cisplatino/farmacología , Humanos , Inflamación/tratamiento farmacológico , Neoplasias/inducido químicamente , Neoplasias/tratamiento farmacológico , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Transducción de Señal
4.
N Engl J Med ; 374(23): 2246-55, 2016 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-27276562

RESUMEN

BACKGROUND: Whole-exome sequencing has transformed gene discovery and diagnosis in rare diseases. Translation into disease-modifying treatments is challenging, particularly for intellectual developmental disorder. However, the exception is inborn errors of metabolism, since many of these disorders are responsive to therapy that targets pathophysiological features at the molecular or cellular level. METHODS: To uncover the genetic basis of potentially treatable inborn errors of metabolism, we combined deep clinical phenotyping (the comprehensive characterization of the discrete components of a patient's clinical and biochemical phenotype) with whole-exome sequencing analysis through a semiautomated bioinformatics pipeline in consecutively enrolled patients with intellectual developmental disorder and unexplained metabolic phenotypes. RESULTS: We performed whole-exome sequencing on samples obtained from 47 probands. Of these patients, 6 were excluded, including 1 who withdrew from the study. The remaining 41 probands had been born to predominantly nonconsanguineous parents of European descent. In 37 probands, we identified variants in 2 genes newly implicated in disease, 9 candidate genes, 22 known genes with newly identified phenotypes, and 9 genes with expected phenotypes; in most of the genes, the variants were classified as either pathogenic or probably pathogenic. Complex phenotypes of patients in five families were explained by coexisting monogenic conditions. We obtained a diagnosis in 28 of 41 probands (68%) who were evaluated. A test of a targeted intervention was performed in 18 patients (44%). CONCLUSIONS: Deep phenotyping and whole-exome sequencing in 41 probands with intellectual developmental disorder and unexplained metabolic abnormalities led to a diagnosis in 68%, the identification of 11 candidate genes newly implicated in neurometabolic disease, and a change in treatment beyond genetic counseling in 44%. (Funded by BC Children's Hospital Foundation and others.).


Asunto(s)
Exoma , Pruebas Genéticas/métodos , Errores Innatos del Metabolismo/genética , Análisis de Secuencia de ADN/métodos , Adolescente , Adulto , Niño , Preescolar , Femenino , Genotipo , Humanos , Lactante , Discapacidad Intelectual/genética , Masculino , Errores Innatos del Metabolismo/diagnóstico , Fenotipo , Adulto Joven
5.
Pediatr Blood Cancer ; 65(3)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29271558

RESUMEN

BACKGROUND: Anthracyclines are a class of highly effective chemotherapeutic drugs commonly used to treat cancer patients. Anthracyclines, however, are associated with the development of serious adverse reactions, including anthracycline-induced cardiotoxicity (ACT). It is not possible, within current practice, to accurately individualize treatment to minimize risk. PROCEDURE: Recently, genetic variants have been associated with the risk of ACT in children. Building on these findings and the related genetic test, a predictive model was developed which classifies pediatric patients by their risk of developing ACT. We assessed the value of this ACT-predictive risk classification in addressing ACT. RESULTS: With current care, the estimated average lifetime cost of ACT is $8,667 per anthracycline-treated patient and approximately 7% of patients are expected to die from ACT. The projected impact of the information from the new predictive model is a 17% reduction in the risk of mortality from ACT and savings of about 6%: lives saved and lower costs. CONCLUSION: The newly identified genetic variants associated with the risk of ACT provide information that allows a more reliable prediction of the risk of ACT for a given patient and can be obtained at a very moderate cost, which is expected to lead to meaningful progress in reducing harm and costs associated with ACT.


Asunto(s)
Antraciclinas/efectos adversos , Cardiotoxicidad , Neoplasias/tratamiento farmacológico , Pruebas de Farmacogenómica/economía , Pruebas de Farmacogenómica/métodos , Antineoplásicos/efectos adversos , Niño , Análisis Costo-Beneficio , Árboles de Decisión , Femenino , Costos de la Atención en Salud , Humanos , Masculino
6.
Am J Hum Genet ; 94(3): 453-61, 2014 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-24530203

RESUMEN

Four children in three unrelated families (one consanguineous) presented with lethargy, hyperlactatemia, and hyperammonemia of unexplained origin during the neonatal period and early childhood. We identified and validated three different CA5A alterations, including a homozygous missense mutation (c.697T>C) in two siblings, a homozygous splice site mutation (c.555G>A) leading to skipping of exon 4, and a homozygous 4 kb deletion of exon 6. The deleterious nature of the homozygous mutation c.697T>C (p.Ser233Pro) was demonstrated by reduced enzymatic activity and increased temperature sensitivity. Carbonic anhydrase VA (CA-VA) was absent in liver in the child with the homozygous exon 6 deletion. The metabolite profiles in the affected individuals fit CA-VA deficiency, showing evidence of impaired provision of bicarbonate to the four enzymes that participate in key pathways in intermediary metabolism: carbamoylphosphate synthetase 1 (urea cycle), pyruvate carboxylase (anaplerosis, gluconeogenesis), propionyl-CoA carboxylase, and 3-methylcrotonyl-CoA carboxylase (branched chain amino acids catabolism). In the three children who were administered carglumic acid, hyperammonemia resolved. CA-VA deficiency should therefore be added to urea cycle defects, organic acidurias, and pyruvate carboxylase deficiency as a treatable condition in the differential diagnosis of hyperammonemia in the neonate and young child.


Asunto(s)
Anhidrasa Carbónica V/deficiencia , Anhidrasa Carbónica V/genética , Hiperamonemia/genética , Adolescente , Secuencia de Bases , Niño , Preescolar , Exones , Femenino , Eliminación de Gen , Variación Genética , Homocigoto , Humanos , Hiperamonemia/terapia , Lactante , Hígado/enzimología , Masculino , Datos de Secuencia Molecular , Mutación Missense , Linaje , Análisis de Secuencia de ADN , Temperatura
7.
Br J Clin Pharmacol ; 82(3): 683-95, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27197003

RESUMEN

AIMS: Anthracycline-induced cardiotoxicity (ACT) occurs in 57% of treated patients and remains an important limitation of anthracycline-based chemotherapy. In various genetic association studies, potential genetic risk markers for ACT have been identified. Therefore, we developed evidence-based clinical practice recommendations for pharmacogenomic testing to further individualize therapy based on ACT risk. METHODS: We followed a standard guideline development process, including a systematic literature search, evidence synthesis and critical appraisal, and the development of clinical practice recommendations with an international expert group. RESULTS: RARG rs2229774, SLC28A3 rs7853758 and UGT1A6 rs17863783 variants currently have the strongest and the most consistent evidence for association with ACT. Genetic variants in ABCC1, ABCC2, ABCC5, ABCB1, ABCB4, CBR3, RAC2, NCF4, CYBA, GSTP1, CAT, SULT2B1, POR, HAS3, SLC22A7, SCL22A17, HFE and NOS3 have also been associated with ACT, but require additional validation. We recommend pharmacogenomic testing for the RARG rs2229774 (S427L), SLC28A3 rs7853758 (L461L) and UGT1A6*4 rs17863783 (V209V) variants in childhood cancer patients with an indication for doxorubicin or daunorubicin therapy (Level B - moderate). Based on an overall risk stratification, taking into account genetic and clinical risk factors, we recommend a number of management options including increased frequency of echocardiogram monitoring, follow-up, as well as therapeutic options within the current standard of clinical practice. CONCLUSIONS: Existing evidence demonstrates that genetic factors have the potential to improve the discrimination between individuals at higher and lower risk of ACT. Genetic testing may therefore support both patient care decisions and evidence development for an improved prevention of ACT.


Asunto(s)
Antraciclinas/efectos adversos , Cardiotoxicidad/prevención & control , Pruebas Genéticas , Medicina Basada en la Evidencia , Predisposición Genética a la Enfermedad/genética , Humanos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Factores de Riesgo
8.
Ther Drug Monit ; 38(4): 423-31, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26960170

RESUMEN

Currently no pharmacogenomics-based criteria exist to guide clinicians in identifying individuals who are at risk of hearing loss from cisplatin-based chemotherapy. This review summarizes findings from pharmacogenomic studies that report genetic polymorphisms associated with cisplatin-induced hearing loss and aims to (1) provide up-to-date information on new developments in the field, (2) provide recommendations for the use of pharmacogenetic testing in the prevention, assessment, and management of cisplatin-induced hearing loss in children and adults, and (3) identify knowledge gaps to direct and prioritize future research. These practice recommendations for pharmacogenetic testing in the context of cisplatin-induced hearing loss reflect a review and evaluation of recent literature, and are designed to assist clinicians in providing optimal clinical care for patients receiving cisplatin-based chemotherapy.


Asunto(s)
Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Cisplatino/efectos adversos , Cisplatino/uso terapéutico , Marcadores Genéticos/genética , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/genética , Humanos , Farmacogenética/métodos , Polimorfismo Genético/genética
9.
PLoS Pathog ; 9(7): e1003518, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23935490

RESUMEN

To further its pathogenesis, S. Typhimurium delivers effector proteins into host cells, including the novel E3 ubiquitin ligase (NEL) effector SspH2. Using model systems in a cross-kingdom approach we gained further insight into the molecular function of this effector. Here, we show that SspH2 modulates innate immunity in both mammalian and plant cells. In mammalian cell culture, SspH2 significantly enhanced Nod1-mediated IL-8 secretion when transiently expressed or bacterially delivered. In addition, SspH2 also enhanced an Rx-dependent hypersensitive response in planta. In both of these nucleotide-binding leucine rich repeat receptor (NLR) model systems, SspH2-mediated phenotypes required its catalytic E3 ubiquitin ligase activity and interaction with the conserved host protein SGT1. SGT1 has an essential cell cycle function and an additional function as an NLR co-chaperone in animal and plant cells. Interaction between SspH2 and SGT1 was restricted to SGT1 proteins that have NLR co-chaperone function and accordingly, SspH2 did not affect SGT1 cell cycle functions. Mechanistic studies revealed that SspH2 interacted with, and ubiquitinated Nod1 and could induce Nod1 activity in an agonist-independent manner if catalytically active. Interestingly, SspH2 in vitro ubiquitination activity and protein stability were enhanced by SGT1. Overall, this work adds to our understanding of the sophisticated mechanisms used by bacterial effectors to co-opt host pathways by demonstrating that SspH2 can subvert immune responses by selectively exploiting the functions of a conserved host co-chaperone.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Inmunidad Innata , Proteínas de la Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas Adaptadoras de Señalización NOD/metabolismo , Salmonella typhimurium/inmunología , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas de Ciclo Celular/química , Línea Celular , Eliminación de Gen , Interacciones Huésped-Patógeno , Humanos , Interleucina-8/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas Mutantes/metabolismo , Inmunidad de la Planta , Hojas de la Planta/genética , Hojas de la Planta/inmunología , Hojas de la Planta/metabolismo , Hojas de la Planta/microbiología , Proteínas de Plantas/metabolismo , Plantas Modificadas Genéticamente , Estabilidad Proteica , Proteínas Recombinantes/metabolismo , Salmonella typhimurium/metabolismo , Nicotiana/genética , Nicotiana/inmunología , Nicotiana/metabolismo , Nicotiana/microbiología , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Regulación hacia Arriba
10.
Mol Cell Proteomics ; 12(6): 1632-43, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23459991

RESUMEN

During the late stages of infection, Salmonella secretes numerous effectors through a type III secretion system that is encoded within Salmonella pathogenicity island 2 (SPI2). Despite the importance of SPI2 as a major virulence factor leading to the systemic spread of the bacteria and diseases, a global view of its effects on host responses is still lacking. Here, we measured global impacts of SPI2 effectors on the host phosphorylation and protein expression levels in RAW264.7 and in HeLa cells, as macrophage and nonphagocytic models of infection. We observe that SPI2 effectors differentially modulate the host phosphoproteome and cellular processes (e.g. protein trafficking, cytoskeletal regulation, and immune signaling) in a host cell-dependent manner. Our unbiased approach reveals the involvement of many previously unrecognized proteins, including E3 ligases (HERC4, RanBP2, and RAD18), kinases (CDK, SIK3, and WNK1), and histones (H2B1F, H4, and H15), in late stages of Salmonella infection. Furthermore, from this phosphoproteome analysis and other quantitative screens, we identified HSP27 as a direct in vitro and in vivo molecular target of the only type III secreted kinase, SteC. Using biochemical and cell biological assays, we demonstrate that SteC phosphorylates multiple sites in HSP27 and induces actin rearrangement through this protein. Together, these results provide a broader landscape of host players contributing to specific processes/pathways mediated by SPI2 effectors than was previously appreciated.


Asunto(s)
Proteínas Bacterianas/genética , Islas Genómicas , Proteínas de Choque Térmico HSP27/genética , Macrófagos/metabolismo , Fosfoproteínas/genética , Proteínas Quinasas/genética , Proteoma/genética , Salmonella typhimurium/genética , Salmonella typhimurium/patogenicidad , Actinas/genética , Actinas/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Línea Celular , Regulación de la Expresión Génica , Proteínas de Choque Térmico HSP27/metabolismo , Células HeLa , Proteínas de Choque Térmico , Interacciones Huésped-Patógeno , Humanos , Macrófagos/citología , Macrófagos/microbiología , Ratones , Chaperonas Moleculares , Fosfoproteínas/metabolismo , Mapeo de Interacción de Proteínas , Proteínas Quinasas/metabolismo , Proteoma/aislamiento & purificación , Proteoma/metabolismo , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
11.
Nature ; 449(7164): 827-34, 2007 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-17943119

RESUMEN

Bacterial pathogens operate by attacking crucial intracellular pathways in their hosts. These pathogens usually target more than one intracellular pathway and often interact at several points in each of these pathways to commandeer them fully. Although different bacterial pathogens tend to exploit similar pathway components in the host, the way in which they 'hijack' host cells usually differs. Knowledge of how pathogens target distinct cytoskeletal components and immune-cell signalling pathways is rapidly advancing, together with the understanding of bacterial virulence at a molecular level. Studying how these bacterial pathogens subvert host-cell pathways is central to understanding infectious disease.


Asunto(s)
Bacterias/patogenicidad , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/patología , Interacciones Huésped-Patógeno , Animales , Presentación de Antígeno , Citoesqueleto/metabolismo , Humanos , Transducción de Señal
12.
J Biol Chem ; 286(27): 24023-35, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21566117

RESUMEN

Gram-negative bacterial pathogens have developed specialized secretion systems to transfer bacterial proteins directly into host cells. These bacterial effectors are central to virulence and reprogram host cell processes to favor bacterial survival, colonization, and proliferation. Knowing the complete set of effectors encoded by a particular pathogen is the key to understanding bacterial disease. In addition, the identification of the molecular assemblies that these effectors engage once inside the host cell is critical to determining the mechanism of action of each effector. In this work we used stable isotope labeling of amino acids in cell culture (SILAC), a powerful quantitative proteomics technique, to identify the proteins secreted by the Salmonella pathogenicity island-2 type three secretion system (SPI-2 T3SS) and to characterize the host interaction partners of SPI-2 effectors. We confirmed many of the known SPI-2 effectors and were able to identify several novel substrate candidates of this secretion system. We verified previously published host protein-effector binding pairs and obtained 11 novel interactions, three of which were investigated further and confirmed by reciprocal co-immunoprecipitation. The host cell interaction partners identified here suggest that Salmonella SPI-2 effectors target, in a concerted fashion, cellular processes such as cell attachment and cell cycle control that are underappreciated in the context of infection. The technology outlined in this study is specific and sensitive and serves as a robust tool for the identification of effectors and their host targets that is readily amenable to the study of other bacterial pathogens.


Asunto(s)
Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos/fisiología , Islas Genómicas/fisiología , Interacciones Huésped-Patógeno/fisiología , Proteínas de la Membrana/metabolismo , Salmonella typhimurium/fisiología , Salmonella typhimurium/patogenicidad , Proteínas Bacterianas/genética , Humanos , Proteínas de la Membrana/genética
13.
Cell Microbiol ; 13(10): 1542-57, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21812888

RESUMEN

Enteropathogenic and enterohaemorrhagic Escherichia coli (EPEC and EHEC respectively) are attaching and effacing bacterial pathogens that cause devastating diarrhoeal disease worldwide. These pathogens depend on a type III secretion system, which functions as a molecular syringe to translocate bacterial effector proteins directly into infected host cells. One of these effectors, NleC, was recently described as a zinc metalloprotease that targets NF-κB Rel-A (p65) and thus contributes to dampening of inflammatory signalling during EPEC and EHEC infection. We have identified the acetyltransferase p300 as an additional target of NleC. Several biochemical techniques were employed to demonstrate specific binding of p300 by NleC. We also show that NleC causes decreased abundance of p300 in cellular nuclei and that the metalloprotease domain of NleC is responsible for this phenotype. Furthermore, we demonstrate that overexpression of p300 can antagonize repression of IL-8 secretion by EPEC and that siRNA knock-down of p300 dampens IL-8 secretion by EPEC ΔnleC-infected cells. We have therefore identified a second target of NleC and provided the first example of a bacterial virulence factor targeting the acetyltransferase p300.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Escherichia coli/patogenicidad , Interacciones Huésped-Patógeno , Factores de Virulencia/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Línea Celular , Humanos , Unión Proteica , Proteolisis
15.
PLoS One ; 17(11): e0276541, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36331922

RESUMEN

The effectiveness of anthracycline chemotherapeutics (e.g., doxorubicin) is limited by anthracycline-induced cardiotoxicity (ACT). A nonsynonymous variant (S427L) in the retinoic acid receptor-γ (RARG) gene has been associated with ACT. This variant causes reduced RARG activity, which is hypothesized to lead to increased susceptibility to ACT through reduced activation of the retinoic acid pathway. This study explored the effects of activating the retinoic acid pathway using a RAR-agonist, all-trans retinoic acid (ATRA), in human cardiomyocytes and mice treated with doxorubicin. In human cardiomyocytes, ATRA induced the gene expression of RARs (RARG, RARB) and repressed the expression of topoisomerase II enzyme genes (TOP2A, TOP2B), which encode for the molecular targets of anthracyclines and repressed downstream ACT response genes. Importantly, ATRA enhanced cell survival of human cardiomyocytes exposed to doxorubicin. The protective effect of ATRA was also observed in a mouse model (B6C3F1/J) of ACT, in which ATRA treatment improved heart function compared to doxorubicin-only treated mice. Histological analyses of the heart also indicated that ATRA treatment reduced the pathology associated with ACT. These findings provide additional evidence for the retinoic acid pathway's role in ACT and suggest that the RAR activator ATRA can modulate this pathway to reduce ACT.


Asunto(s)
Antraciclinas , Cardiotoxicidad , Animales , Humanos , Ratones , Antraciclinas/toxicidad , Antibióticos Antineoplásicos/farmacología , Cardiotoxicidad/genética , Cardiotoxicidad/prevención & control , Cardiotoxicidad/metabolismo , ADN-Topoisomerasas de Tipo II/genética , ADN-Topoisomerasas de Tipo II/metabolismo , Doxorrubicina/toxicidad , Doxorrubicina/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Inhibidores de Topoisomerasa II/farmacología , Tretinoina/farmacología , Tretinoina/metabolismo , Receptor de Ácido Retinoico gamma
16.
NPJ Genom Med ; 6(1): 35, 2021 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-34021165

RESUMEN

Anthracyclines are highly effective chemotherapeutic agents; however, their clinical utility is limited by severe anthracycline-induced cardiotoxicity (ACT). Genome-wide association studies (GWAS) have uncovered several genetic variants associated with ACT, but the impact of these findings requires further elucidation. We conducted a transcriptome-wide association study (TWAS) using our previous GWAS summary statistics (n = 280 patients) to identify gene expression-related associations with ACT. We identified a genetic association between decreased expression of GDF5 and ACT (Z-score = -4.30, P = 1.70 × 10-5), which was replicated in an independent cohort (n = 845 patients, P = 3.54 × 10-3). Additionally, cell viability of GDF5-silenced human cardiac myocytes was significantly decreased in response to anthracycline treatment. Subsequent gene set enrichment and pathway analyses of the TWAS data revealed that genes essential for survival, cardioprotection and response to anthracyclines, as well as genes involved in ribosomal, spliceosomal and cardiomyopathy pathways are important for the development of ACT.

17.
Clin Pharmacol Ther ; 106(2): 350-359, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31012503

RESUMEN

Cisplatin is a highly effective chemotherapeutic. Unfortunately, its use is limited by cisplatin-induced ototoxicity (CIO). Substantial research has been performed to uncover the genetic variants associated with CIO; however, there has been a lack of consistency in the results that have been reported. This paper aims to provide an overview of the current state of CIO genomics research, delving into the shortcomings of past research, and providing recommendations for future avenues of study.


Asunto(s)
Cisplatino/farmacología , Ototoxicidad , Farmacogenética , Antineoplásicos/farmacología , Humanos , Ototoxicidad/etiología , Ototoxicidad/genética
18.
Nat Genet ; 50(8): 1081-1085, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30013178

RESUMEN

Multiple sclerosis (MS) is a disease of the central nervous system treated with disease-modifying therapies, including the biologic, interferon-ß (IFN-ß). Up to 60% of IFN-ß-exposed MS patients develop abnormal biochemical liver test results1,2, and 1 in 50 experiences drug-induced liver injury3. Since genomic variation contributes to other forms of drug-induced liver injury4,5, we aimed to identify biomarkers of IFN-ß-induced liver injury using a two-stage genome-wide association study. The rs2205986 variant, previously linked to differential expression of IRF6, surpassed genome-wide significance in the combined two-stage analysis (P = 2.3 × 10-8, odds ratio = 8.3, 95% confidence interval = 3.6-19.2). Analysis of an independent cohort of IFN-ß-treated MS patients identified via electronic medical records showed that rs2205986 was also associated with increased peak levels of aspartate aminotransferase (P = 7.6 × 10-5) and alkaline phosphatase (P = 4.9 × 10-4). We show that these findings may be applicable to predicting IFN-ß-induced liver injury, offering insight into its safer use.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Variación Genética/genética , Factores Reguladores del Interferón/genética , Interferón beta/genética , Esclerosis Múltiple/genética , Femenino , Estudio de Asociación del Genoma Completo/métodos , Humanos , Masculino
19.
PLoS One ; 12(4): e0175711, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28406961

RESUMEN

Cisplatin is a highly-effective and widely-used chemotherapeutic agent that causes ototoxicity in many patients. Pharmacogenomic studies of key genes controlling drug biotransformation identified variants in thiopurine methyltransferase (TPMT) as predictors of cisplatin-induced ototoxicity, although the mechanistic basis of this interaction has not been reported. Expression constructs of TPMT*3A, *3B and *3C variants were generated and monitored in cultured cells. Cellular TPMT*3A levels were detected at >20-fold lower amounts than the wild type confirming the unstable nature of this variant. The expression of wild type TPMT (TPMT*1) in two murine ear cell lines, HEI-OC1 and UB/OC-1, significantly mitigated their susceptibility to cisplatin toxicity. Cisplatin treatment induced Tlr4 gene expression in HEI-OC1 cells and this response was blunted by the expression of wild type TPMT but not TPMT*3A. In line with the significant mitigation of TPMT*1-expressing cells to cisplatin cytotoxicity, these findings demonstrate a drug-gene interaction between increased TPMT activity and decreased susceptibility to cisplatin-induced toxicity of inner ear cells.


Asunto(s)
Cisplatino/efectos adversos , Oído Interno/citología , Células Ciliadas Auditivas Internas/efectos de los fármacos , Metiltransferasas/genética , Variantes Farmacogenómicas , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Oído Interno/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Células Ciliadas Auditivas Internas/citología , Haplotipos , Humanos , Ratones , Receptor Toll-Like 4/genética
20.
JAMA Oncol ; 3(11): 1558-1562, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28448657

RESUMEN

IMPORTANCE: Cisplatin-induced ototoxic effects are an important complication that affects testicular cancer survivors as a consequence of treatment. The identification of genetic variants associated with this adverse drug reaction will further our mechanistic understanding of its development and potentially lead to strategies to prevent ototoxic effects. OBJECTIVE: To identify the genetic variants associated with cisplatin-induced ototoxic effects in adult testicular cancer patients. DESIGN, SETTING, AND PARTICIPANTS: This retrospective study was performed by the Canadian Pharmacogenomics Network for Drug Safety using patients recruited from 5 adult oncology treatment centers across Canada. Male patients who were 17 years or older, diagnosed with germ cell testicular cancer, and previously treated with cisplatin-based chemotherapy were recruited from July 2009 to April 2013 using active surveillance methodology. Cisplatin-induced ototoxic effects were independently diagnosed by 2 audiologists. Patients were genotyped for 7907 variants using a custom pharmacogenomic array. Logistic regression was used to identify genetic variants that were significantly associated with ototoxic effects. The validity of these findings was confirmed through independent replication and cell-based functional assays. EXPOSURES: Cisplatin-based chemotherapy. MAIN OUTCOMES AND MEASURES: Cisplatin-induced ototoxic effects. RESULTS: After exclusions, 188 patients (median [interquartile range] age, 31 [24-39] years) were enrolled in this study to form the discovery and replication cohorts. Association and fine-mapping analyses identified a protein-coding variant, rs4788863 in SLC16A5, that was associated with protection against cisplatin-induced ototoxic effects in 2 independent cohorts (combined cohort: odds ratio, 0.06; 95% CI, 0.02-0.22; P = 2.17 × 10-7). Functional validation of this transporter gene revealed that in vitro SLC16A5-silencing altered cellular responses to cisplatin treatment, supporting a role for SLC16A5 in the development of cisplatin-induced ototoxic effects. These results were further supported by the literature, which provided confirmatory evidence for the role that SLC16A5 plays in hearing. CONCLUSIONS AND RELEVANCE: This study has identified a novel association between protein-coding variation in SLC16A5 and cisplatin-induced ototoxic effects. These findings have provided insight into the molecular mechanisms of this adverse drug reaction in adult patients with germ cell testicular cancer. Given that previous studies have shown that cimetidine, an SLC16A5-inhibitor, prevents murine cisplatin-induced ototoxic effects, the findings from this study have important implications for otoprotectant strategies in humans.


Asunto(s)
Antineoplásicos/efectos adversos , Cisplatino/efectos adversos , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/genética , Transportadores de Ácidos Monocarboxílicos/genética , Variantes Farmacogenómicas , Neoplasias Testiculares/tratamiento farmacológico , Adolescente , Adulto , Canadá , Relación Dosis-Respuesta a Droga , Predisposición Genética a la Enfermedad , Células HeLa , Pérdida Auditiva/diagnóstico , Pérdida Auditiva/metabolismo , Humanos , Modelos Logísticos , Masculino , Transportadores de Ácidos Monocarboxílicos/efectos de los fármacos , Transportadores de Ácidos Monocarboxílicos/metabolismo , Farmacogenética , Pruebas de Farmacogenómica , Fenotipo , Interferencia de ARN , Estudios Retrospectivos , Factores de Riesgo , Transfección , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA