Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 165
Filtrar
1.
PLoS Med ; 19(2): e1003897, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35113855

RESUMEN

BACKGROUND: Epidemiological studies have reported conflicting findings on the potential adverse effects of long-term antihypertensive medication use on cancer risk. Naturally occurring variation in genes encoding antihypertensive drug targets can be used as proxies for these targets to examine the effect of their long-term therapeutic inhibition on disease outcomes. METHODS AND FINDINGS: We performed a mendelian randomization analysis to examine the association between genetically proxied inhibition of 3 antihypertensive drug targets and risk of 4 common cancers (breast, colorectal, lung, and prostate). Single-nucleotide polymorphisms (SNPs) in ACE, ADRB1, and SLC12A3 associated (P < 5.0 × 10-8) with systolic blood pressure (SBP) in genome-wide association studies (GWAS) were used to proxy inhibition of angiotensin-converting enzyme (ACE), ß-1 adrenergic receptor (ADRB1), and sodium-chloride symporter (NCC), respectively. Summary genetic association estimates for these SNPs were obtained from GWAS consortia for the following cancers: breast (122,977 cases, 105,974 controls), colorectal (58,221 cases, 67,694 controls), lung (29,266 cases, 56,450 controls), and prostate (79,148 cases, 61,106 controls). Replication analyses were performed in the FinnGen consortium (1,573 colorectal cancer cases, 120,006 controls). Cancer GWAS and FinnGen consortia data were restricted to individuals of European ancestry. Inverse-variance weighted random-effects models were used to examine associations between genetically proxied inhibition of these drug targets and risk of cancer. Multivariable mendelian randomization and colocalization analyses were employed to examine robustness of findings to violations of mendelian randomization assumptions. Genetically proxied ACE inhibition equivalent to a 1-mm Hg reduction in SBP was associated with increased odds of colorectal cancer (odds ratio (OR) 1.13, 95% CI 1.06 to 1.22; P = 3.6 × 10-4). This finding was replicated in the FinnGen consortium (OR 1.40, 95% CI 1.02 to 1.92; P = 0.035). There was little evidence of association of genetically proxied ACE inhibition with risk of breast cancer (OR 0.98, 95% CI 0.94 to 1.02, P = 0.35), lung cancer (OR 1.01, 95% CI 0.92 to 1.10; P = 0.93), or prostate cancer (OR 1.06, 95% CI 0.99 to 1.13; P = 0.08). Genetically proxied inhibition of ADRB1 and NCC were not associated with risk of these cancers. The primary limitations of this analysis include the modest statistical power for analyses of drug targets in relation to some less common histological subtypes of cancers examined and the restriction of the majority of analyses to participants of European ancestry. CONCLUSIONS: In this study, we observed that genetically proxied long-term ACE inhibition was associated with an increased risk of colorectal cancer, warranting comprehensive evaluation of the safety profiles of ACE inhibitors in clinical trials with adequate follow-up. There was little evidence to support associations across other drug target-cancer risk analyses, consistent with findings from short-term randomized controlled trials for these medications.


Asunto(s)
Antihipertensivos/efectos adversos , Análisis de la Aleatorización Mendeliana/métodos , Neoplasias/genética , Peptidil-Dipeptidasa A/genética , Receptores Adrenérgicos beta 1/genética , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Femenino , Estudio de Asociación del Genoma Completo/métodos , Humanos , Masculino , Neoplasias/inducido químicamente , Neoplasias/epidemiología , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo , Miembro 3 de la Familia de Transportadores de Soluto 12/genética
2.
Toxicol Ind Health ; 37(12): 727-736, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34730462

RESUMEN

Epidemiological evidence suggests that the metabolic profiles of each individual exposed to arsenic (As) are related to the risk of cancer, coronary heart disease, and diabetes. The arsenite methyltransferase (AS3MT) gene plays a key role in As metabolism. Several single nucleotide polymorphisms in the AS3MT gene may affect both enzyme activity and gene transcription. AS3MT polymorphisms are associated with the proportions of monomethylarsenic acid (MMA) and dimethylarsenic acid (DMA) in urine as well as the incidence of cancer. P21 protein is a cyclin-dependent kinase inhibitor. Mutations of the P21 gene have been found in cancer patients. In our study, we investigate whether polymorphisms of the AS3MT gene alter As methylation capacity and adversely affect the P21 gene in arsenic trioxide plant workers. The DNA damage was examined by the quantitative polymerase chain reaction. Restriction fragment length polymorphism was used to analyze the genotype of the AS3MT gene. The results showed that DNA damage in P21 gene fragments was greater in those individuals exposed to high levels of As. There was a strong positive correlation between the DNA damage to P21 gene fragments and the percentage of MMA in urine. However, DNA damage in P21 gene fragments was negatively associated with the percentage of DMA in urine (%uDMA), primary methylation index (PMI), and secondary methylation index. We found that subjects with the rs7085104 GG or GA allele were associated with higher %uDMA and PMI and less DNA damage. The subjects with the rs11191454 GG+GA or GA allele were also associated with higher %uDMA and PMI and less DNA damage. Our results suggest that rs1191454 and rs7085104 in the AS3MT gene affect the As-induced DNA damage by altering individual metabolic efficiency.


Asunto(s)
Trióxido de Arsénico/efectos adversos , Trióxido de Arsénico/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Metiltransferasas/genética , Exposición Profesional/efectos adversos , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Adulto , Alelos , Arsénico , China , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Femenino , Humanos , Masculino , Metiltransferasas/orina , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa
3.
Ann Hematol ; 100(10): 2453-2462, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34269838

RESUMEN

BACKGROUND: Primary immune thrombocytopenia (ITP) is an autoimmune disorder characterized by decreased platelet count. While corticosteroids are a useful first-line therapy for ITP patients, their long-term effectiveness is limited, and the determinants of corticosteroid sensitivity in ITP patients remain largely unknown. Sirtuin 1 (SIRT1), a member of the mammalian sirtuin family, is related to the anti-inflammatory effects of corticosteroids. Here, we investigate the contribution of the SIRT1 single-nucleotide polymorphisms (SNPs) rs12778366 and rs4746720 to ITP susceptibility. METHODS: We recruited 330 ITP patients and 309 healthy controls from Han population, and performed genotyping of SIRT1 rs12778366 and rs4746720 using a MassARRAY system. The results were validated in another 55 ITP patients from ethnic minorities. RESULTS: Using clinical data of patients and controls from Han polulation, including corticosteroid sensitivity, susceptibility, refractoriness, and severity, our results revealed that the CC/TC genotypes of SIRT1 rs12778366 were associated with a 2.034-fold increased risk of corticosteroid resistance compared to the homozygous major TT genotype (dominant, CC/TC vs. TT, OR = 2.034, 95% CI = 1.039-3.984, p = 0.038). In contrast, the CC/CT genotype of SIRT1 rs4746720 showed a 0.560-fold decreased risk of corticosteroid resistance (dominant, 95% CI = 0.321-0.976, OR = 0.560, p = 0.041). The C allele substitute in SIRT1 rs12778366 was significantly associated with the corticosteroid sensitivity of ITP patients (p = 0.021). The similar results were obtained in minority ITP patients. CONCLUSION: This study indicates that SIRT1 rs12778366 and rs4746720 may be genetic factors related to corticosteroid sensitivity in ITP patients.


Asunto(s)
Corticoesteroides/uso terapéutico , Polimorfismo de Nucleótido Simple , Púrpura Trombocitopénica Idiopática/tratamiento farmacológico , Sirtuina 1/genética , Corticoesteroides/farmacología , Adulto , Resistencia a Medicamentos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Púrpura Trombocitopénica Idiopática/genética , Resultado del Tratamiento
4.
Int J Mol Sci ; 22(14)2021 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-34298919

RESUMEN

This study was conducted to investigate doubled haploid (DH) lines produced between high GSL (HGSL) Brassica rapa ssp. trilocularis (yellow sarson) and low GSL (LGSL) B. rapa ssp. chinensis (pak choi) parents. In total, 161 DH lines were generated. GSL content of HGSL DH lines ranged from 44.12 to 57.04 µmol·g-1·dry weight (dw), which is within the level of high GSL B. rapa ssp. trilocularis (47.46 to 59.56 µmol g-1 dw). We resequenced five of the HGSL DH lines and three of the LGSL DH lines. Recombination blocks were formed between the parental and DH lines with 108,328 single-nucleotide polymorphisms in all chromosomes. In the measured GSL, gluconapin occurred as the major substrate in HGSL DH lines. Among the HGSL DH lines, BrYSP_DH005 had glucoraphanin levels approximately 12-fold higher than those of the HGSL mother plant. The hydrolysis capacity of GSL was analyzed in HGSL DH lines with a Korean pak choi cultivar as a control. Bioactive compounds, such as 3-butenyl isothiocyanate, 4-pentenyl isothiocyanate, 2-phenethyl isothiocyanate, and sulforaphane, were present in the HGSL DH lines at 3-fold to 6.3-fold higher levels compared to the commercial cultivar. The selected HGSL DH lines, resequencing data, and SNP identification were utilized for genome-assisted selection to develop elite GSL-enriched cultivars and the industrial production of potential anti-cancerous metabolites such as gluconapin and glucoraphanin.


Asunto(s)
Brassica rapa/genética , Glucosinolatos/genética , Brassica rapa/efectos de los fármacos , Genotipo , Glucosinolatos/farmacología , Haploidia , Isotiocianatos/farmacología , Oximas/farmacología , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Polimorfismo de Nucleótido Simple/genética , Sulfóxidos/farmacología
5.
Int J Mol Sci ; 22(14)2021 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-34299222

RESUMEN

FMS-like tyrosine kinase 3 (FLT3) gene mutations have been found in more than one-third of Acute Myeloid Leukemia (AML) cases. The most common point mutation in FLT3 occurs at the 835th residue (D835A/E/F/G/H/I/N/V/Y), in the activation loop region. The D835 residue is critical in maintaining FLT3 inactive conformation; these mutations might influence the interaction with clinically approved AML inhibitors used to treat the AML. The molecular mechanism of each of these mutations and their interactions with AML inhibitors at the atomic level is still unknown. In this manuscript, we have investigated the structural consequence of native and mutant FLT-3 proteins and their molecular mechanisms at the atomic level, using molecular dynamics simulations (MDS). In addition, we use the molecular docking method to investigate the binding pattern between the FLT-3 protein and AML inhibitors upon mutations. This study apparently elucidates that, due to mutations in the D835, the FLT-3 structure loses its conformation and becomes more flexible compared to the native FLT3 protein. These structural changes are suggested to contribute to the relapse and resistance responses to AML inhibitors. Identifying the effects of FLT3 at the molecular level will aid in developing a personalized therapeutic strategy for treating patients with FLT-3-associated AML.


Asunto(s)
Leucemia Mieloide Aguda/genética , Tirosina Quinasa 3 Similar a fms/genética , Simulación por Computador , Resistencia a Antineoplásicos/genética , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Simulación del Acoplamiento Molecular/métodos , Simulación de Dinámica Molecular , Mutación/efectos de los fármacos , Mutación/genética , Mutación Puntual/efectos de los fármacos , Mutación Puntual/genética , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Polimorfismo de Nucleótido Simple/genética , Conformación Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Tirosina Quinasa 3 Similar a fms/metabolismo
6.
Sci Rep ; 11(1): 5449, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33750834

RESUMEN

Response to anti-TNF therapy is of pivotal importance in patients with Crohn's disease (CD). Here we integrated our and previously reported PBMC derived transcriptomic and genomic data for identification of biomarkers for discrimination between responders and non-responders to anti-TNF therapy. CD patients, who were naïve with respect to the treatment with biologicals, were enrolled in the study. DNA and RNA were extracted from peripheral blood mononuclear cells. RNA-seq was performed using BGISEQ-500. Genotyping was performed using Infinium Global Screening Array. Association regressions were carried out with 12 week response to adalimumab as an outcome variable. RNA-seq analysis confirmed 7 out of 65 previously suggested genes involved in anti-TNF response. Subsequently, analysis of single nucleotide variants in regions of confirmed genes identified 5 variants near MMD and two in ELOVL7 intronic regions associated with treatment response to anti-TNF. Functional analysis has shown that rs1465352, rs4422035 and rs78620886 are listed at H3K9ac_Pro histone modification epigenetic mark. The present study confirmed MMD and ELOVL7 involvement in anti-TNF response and revealed that the regulation of MMD and ELOVL7 gene regions in ADA response may be a part of a complex interplay extending from genetic to epigenetic and to transcriptomic level.


Asunto(s)
Adalimumab/uso terapéutico , Enfermedad de Crohn/tratamiento farmacológico , Elongasas de Ácidos Grasos/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Inhibidores del Factor de Necrosis Tumoral/uso terapéutico , Adulto , Enfermedad de Crohn/genética , Femenino , Sitios Genéticos/efectos de los fármacos , Genómica , Humanos , Intrones/efectos de los fármacos , Masculino , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Adulto Joven
7.
Sci Rep ; 11(1): 3780, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33580158

RESUMEN

Identifying in advance who is unlikely to respond to a specific antidepressant treatment is crucial to precision medicine efforts. The current work leverages genome-wide genetic variation and machine learning to predict response to the antidepressant citalopram using data from the Sequenced Treatment Alternatives to Relieve Depression (STAR*D) trial (n = 1257 with both valid genomic and outcome data). A confirmatory approach selected 11 SNPs previously reported to predict response to escitalopram in a sample different from the current study. A novel exploratory approach selected SNPs from across the genome using nested cross-validation with elastic net logistic regression with a predominantly lasso penalty (alpha = 0.99). SNPs from each approach were combined with baseline clinical predictors and treatment response outcomes were predicted using a stacked ensemble of gradient boosting decision trees. Using pre-treatment clinical and symptom predictors only, out-of-fold prediction of a novel treatment response definition based on STAR*D treatment guidelines was acceptable, AUC = .659, 95% CI [0.629, 0.689]. The inclusion of SNPs using confirmatory or exploratory selection methods did not improve the out-of-fold prediction of treatment response (AUCs were .662, 95% CI [0.632, 0.692] and .655, 95% CI [0.625, 0.685], respectively). A similar pattern of results were observed for the secondary outcomes of the presence or absence of distressing side effects regardless of treatment response and achieving remission or satisfactory partial response, assuming medication tolerance. In the current study, incorporating SNP variation into prognostic models did not enhance the prediction of citalopram response in the STAR*D sample.


Asunto(s)
Biomarcadores Farmacológicos/análisis , Trastorno Depresivo Mayor/genética , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Antidepresivos/metabolismo , Antidepresivos/uso terapéutico , Área Bajo la Curva , Citalopram/farmacología , Bases de Datos Factuales , Bases de Datos Genéticas , Árboles de Decisión , Depresión/tratamiento farmacológico , Depresión/genética , Trastorno Depresivo Mayor/tratamiento farmacológico , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Variación Genética/genética , Humanos , Modelos Logísticos , Aprendizaje Automático , Polimorfismo de Nucleótido Simple/genética , Pronóstico , Resultado del Tratamiento
8.
Sci Rep ; 11(1): 3249, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33547330

RESUMEN

The PvuII (rs2234693) Single Nucleotide Polymorphism (SNP) in the gene coding for the estrogen receptor-1 (ESR1), has been found associated with outcome in tamoxifen treated patients with early hormone-receptor positive breast cancer. However, it remains unclear whether this SNP is a predictive marker for tamoxifen efficacy or a prognostic marker for breast cancer outcome. The aim of this study was to examine the prognostic potential of this SNP in postmenopausal early breast cancer patients treated with adjuvant exemestane. Dutch postmenopausal patients randomised to 5 years of adjuvant exemestane of whom tissue was available (N = 807) were selected from the Tamoxifen Exemestane Adjuvant Multinational (TEAM) trial database. The SNP rs2234693 in the ESR1 gene was genotyped on DNA from formalin-fixed paraffin embedded (FFPE) tumor tissue using Taqman assays and related to the primary endpoint disease-free survival (DFS) and secondary endpoint overall survival (OS). Survival analyses were performed using Cox regression analysis. In total 805 patients were included in the analyses (median follow up of 5.22 years) and genotypes were obtained in 97% of the samples. The variant T allele of PvuII in ESR1 (rs2234693) was associated with a better DFS (hazard ratio (HR) 0.689, 95% confidence interval (CI) 0.480-0.989, P = 0.044) in univariate analysis only, and a better OS in both univariate (HR 0.616, 95%, CI 0.411-0.923, P = 0.019) and multivariate analyses (HR 0.571, 95% CI 0.380-0.856, P = 0.007), consistent with a prognostic rather than a predictive drug response effect. Variation of PvuII in the ESR1 gene is related to OS in postmenopausal, early HR + breast cancer patients treated with exemestane in the TEAM study. Variation in the ESR1 gene may therefore be a prognostic marker of early breast cancer survival, and warrants further research.


Asunto(s)
Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/genética , Polimorfismo de Nucleótido Simple , Adulto , Alelos , Androstadienos/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/tratamiento farmacológico , Supervivencia sin Enfermedad , Femenino , Humanos , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Pronóstico
9.
Sci Rep ; 11(1): 1155, 2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33441847

RESUMEN

Predicting lithium response prior to treatment could both expedite therapy and avoid exposure to side effects. Since lithium responsiveness may be heritable, its predictability based on genomic data is of interest. We thus evaluate the degree to which lithium response can be predicted with a machine learning (ML) approach using genomic data. Using the largest existing genomic dataset in the lithium response literature (n = 2210 across 14 international sites; 29% responders), we evaluated the degree to which lithium response could be predicted based on 47,465 genotyped single nucleotide polymorphisms using a supervised ML approach. Under appropriate cross-validation procedures, lithium response could be predicted to above-chance levels in two constituent sites (Halifax, Cohen's kappa 0.15, 95% confidence interval, CI [0.07, 0.24]; and Würzburg, kappa 0.2 [0.1, 0.3]). Variants with shared importance in these models showed over-representation of postsynaptic membrane related genes. Lithium response was not predictable in the pooled dataset (kappa 0.02 [- 0.01, 0.04]), although non-trivial performance was achieved within a restricted dataset including only those patients followed prospectively (kappa 0.09 [0.04, 0.14]). Genomic classification of lithium response remains a promising but difficult task. Classification performance could potentially be improved by further harmonization of data collection procedures.


Asunto(s)
Trastorno Bipolar/tratamiento farmacológico , Trastorno Bipolar/genética , Genómica/métodos , Litio/uso terapéutico , Adolescente , Adulto , Trastorno Bipolar/diagnóstico , Femenino , Humanos , Litio/efectos adversos , Litio/farmacología , Aprendizaje Automático , Masculino , Modelos Genéticos , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Pronóstico , Resultado del Tratamiento , Adulto Joven
10.
Int J Lab Hematol ; 43(5): 1000-1008, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33470551

RESUMEN

INTRODUCTION: Glutathione S-transferase (GST) gene deletion or polymorphic sequence variations lead to decreased enzyme activity that influences susceptibility and response to tyrosine kinase inhibitors in chronic myeloid leukemia (CML). We aimed to analyze relation of different GST gene sequence variants with susceptibility and response to Imatinib in CML. MATERIAL AND METHODS: A total of 150 CML cases and equal number of age and gender matched healthy controls were genotyped for five GST polymorphisms by multiplex-PCR and PCR-RFLP techniques. BCR-ABL1 transcripts were quantified by quantitative Real Time PCR (qRT-PCR). RESULTS: GSTT1, GSTO1, and GSTO2 SNPs revealed no association, while as GSTM1null genotype was observed to protect against the development of CML (OR = 0.53, P = .01). GSTP1 variant genotypes AG (OR = 2.1, P = .003) and GG (OR = 5.6, P < .001), significantly associated with increased risk of CML. Combined genotype analysis showed protective impact of GSTT1present /GSTM1null (OR = 0.44, P = .003) while as GSTT1present /GSTP1-GG (OR = 6.92, P < .001) and GSTM1present /GSTP1-GG (OR = 6.33, P < .001), significantly increased CML risk. GSTM1null genotype individually and in combination with GSTT1present associated with superior rate of major molecular response (MMR) and event free survival (EFS) (log-rank P = .029). GSTO2-AG+GG genotype associated with significantly inferior MMR rates at 3, 6, and 12 months. Also, patients with GSTO2-GG genotype showed significantly reduced EFS (log-rank P = .025). Multivariate analysis confirmed GSTM1null as a better (HR:0.19, P = .029) and GSTO2-GG genotype as an independent poor prognostic factor (HR:2.29, P = .037). CONCLUSION: GSTM1null genotype seems to have a better prognostic role while GSTP1 variants significantly increase CML risk. Also, results support a correlation between disease outcome and GSTO2 polymorphism.


Asunto(s)
Antineoplásicos/uso terapéutico , Glutatión Transferasa/genética , Mesilato de Imatinib/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Adulto , Femenino , Eliminación de Gen , Humanos , India/epidemiología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/epidemiología , Masculino , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Medición de Riesgo , Resultado del Tratamiento
11.
Ribeirão Preto; s.n; 2021. 63 p. ilus.
Tesis en Portugués | LILACS, BDENF | ID: biblio-1379602

RESUMEN

Trata-se de uma revisão de escopo que objetivou relatar as evidências científicas reportadas na literatura referentes a polimorfismos genéticos de nucleotídeo único (SNPs) e a sua relação com o desenvolvimento de sintomas depressivos em pessoas com câncer. A pergunta de pesquisa que norteou esta revisão foi: "Quais polimorfismos genéticos de nucleotídeo único estão relacionados ao desenvolvimento de sintomas depressivos em pacientes oncológicos?". Para a sua condução foi utilizado o referencial teórico proposto por Arksey & O'Malley (2005), acrescido de ampliações de Levac, Colquhoun & O'Brien (2010) e Colquhoun, Levac & O'Brien (2014). Realizou-se buscas sistematizadas nas seguintes bases de dados: PubMed, Embase, PsychInfo, Web of Science e Scopus. Encontrou-se 1946 referências únicas e, após rigorosa seleção, 10 artigos atenderam aos três critérios de inclusão propostos: (1) estudos clínicos conduzidos com pacientes oncológicos; (2) apresentar relação entre SNPs e o desenvolvimento de sintomas depressivos; (3) publicados em português, inglês ou espanhol. Dos 10 artigos incluídos na amostra final, 70% tratava-se de estudos longitudinais, a maioria produzidos nos Estados Unidos (40%), com publicação predominante entre os anos 2012-2014 (60%). A população predominantemente estudada foram mulheres acometidas por câncer de mama (60%) e os sintomas depressivos foram, em sua maioria, avaliados por intermédio de instrumentos validados cientificamente (80%). Os SNPs relacionados aos sintomas depressivos foram encontrados, sobretudo, em genes da via inflamatória (6/11, 54,5%), e o polimorfismo mais estudado foi o rs6265 do gene BDNF, da via de transdução de sinais (4/11, 36,3%). Os principais achados dessa revisão, demonstram que portadores do genótipo Met-BDNF tendem a apresentar maior risco de desenvolvimento e agravamento dos sintomas depressivos. Além disso, os SNPs de genes da via da inflamação, especialmente aqueles relacionados à produção de citocinas pró-inflamatórias, podem desempenhar um importante papel preditivo de sintomas depressivos, na referida população. Portanto, pode-se concluir que as evidências disponíveis na literatura apontam que o polimorfismo do gene BDNF Val66Met (rs6265) e SNPs do sistema imunológico, em especial da via inflamatória, despontam como potenciais marcadores biológicos, no contexto do desenvolvimento de sintomas depressivos em pacientes oncológicos


This is a scoping review that aimed to report the scientific evidence reported in the literature regarding single nucleotide genetic polymorphisms (SNPs) and their relation with the development of depressive symptoms in people with cancer. The research question that guided this review was: "Which single nucleotide genetic polymorphisms are related to the development of depressive symptoms in cancer patients?". The theoretical reference proposed by Arksey & O'Malley (2005) was used to conduct it, plus extensions by Levac, Colquhoun & O'Brien (2010) and Colquhoun, Levac & O'Brien (2014). Systematic searches were carried out in the following databases: PubMed, Embase, PsychInfo, Web of Science, and Scopus. 1946 unique references were found and, after rigorous selection, 10 articles met the three proposed inclusion criteria: (1) clinical studies conducted with cancer patients; (2) presenting a relation between SNPs and the development of depressive symptoms; (3) published in Portuguese, English or Spanish. Of the 10 articles included in the final sample, 70% were longitudinal studies, most of them produced in the United States (40%), with a predominant publication between the years 2012-2014 (60%). The predominantly studied population were women affected by breast cancer (60%) and the depressive symptoms were mostly evaluated using scientifically validated instruments (80%). SNPs related to depressive symptoms were found, mainly, in genes of the inflammatory pathway (6/11, 54.5%), and the most studied polymorphism was the rs6265 of the BDNF gene, of the signal transduction pathway (4/11, 36.3%). The main findings of this review demonstrate that patients with the Met-BDNF genotype tend to have a higher risk of developing and worsening depressive symptoms. In addition, the SNPs of genes in the inflammation pathway, especially those related to the production of pro-inflammatory cytokines, can play an important predictive role of depressive symptoms in this population. Therefore, it can be concluded that the evidence available in the literature indicates that the polymorphism of the BDNF gene Val66Met (rs6265) and SNPs of the immune system, especially of the inflammatory pathway, emerge as potential biological markers, in the context of the development of depressive symptoms in oncological patients


Asunto(s)
Biomarcadores , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Depresión , Neoplasias
12.
Theranostics ; 10(21): 9477-9494, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32863940

RESUMEN

Background: Patients with advanced soft tissue sarcomas (STS) have a dismal prognosis with few effective therapeutic options. A defect in the homologous recombination repair (HRR) pathway can accumulate DNA repair errors and gene mutations, which can lead to tumorigenesis. BRCAness describes tumors with an HRR deficiency (HRD) in the absence of a germline BRCA1/2 mutation. However, the characteristics of BRCAness in STS remain largely unknown. Thus, this study aimed to explore the genomic and molecular landscape of BRCAness using whole exome sequencing (WES) in STS, aiming to find a potential target for STS treatment. Methods: WES was performed in 22 STS samples from the First Affiliated Hospital of Sun Yat-sen University to reveal the possible genomic and molecular characteristics. The characteristics were then validated using data of 224 STS samples from The Cancer Genome Atlas (TCGA) database and in vitro data. The analysis of the potential biomarker for BRCAness was performed. Targeted drug susceptibility and combination therapy screening of chemotherapeutics for STS were evaluated in STS cell lines, cell-line-derived xenografts (CDX), and patient-derived xenografts (PDX). Results: Compared with 30 somatic mutation signatures of cancers, high cosine-similarity (0.75) was identified for HRD signatures in the 22 STS samples using nonnegative matrix factorization. Single nucleotide polymorphism indicated a low mutation rate of BRCA1/2 in the 22 STS samples (11.76% and 5.88%, respectively). However, copy number variation analyses demonstrated widespread chromosomal instability; furthermore, 54.55% of STS samples (12/22) carried BRCAness traits. Subsequently, similar genomic and molecular characteristics were also detected in the 224 STS samples from TCGA and in vitro. Poly (ADP-ribose) polymerases (PARP)-1 could be a promising reflection of HRD and therapeutic response. Furthermore, the level of PAR formation was found to be correlated with PARP-1. Subsequently, STS cell lines were determined to be sensitive to PARP inhibitor (PARPi), niraparib. Moreover, based on the screening test of the five common PARPis and combination test among doxorubicin, ifosfamide, dacarbazine, and temozolomide (TMZ), niraparib and TMZ were the most synergistic in STS cell lines. The synergistic effect and safety of niraparib and TMZ combination were also shown in CDX and PDX. Conclusions: BRCAness might be the common genomic and molecular characteristics of majority of STS cases. PARP-1 and PAR could be potential proper and feasible theranostic biomarkers for assessing HRD in patients. STSs were sensitive to PARPi. Moreover, the combination of niraparib and TMZ showed synergistic effect. Niraparib and TMZ could be a promising targeted therapeutic strategy for patients with STS.


Asunto(s)
Indazoles/farmacología , Piperidinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Sarcoma/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Biomarcadores de Tumor/genética , Variaciones en el Número de Copia de ADN/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Poli(ADP-Ribosa) Polimerasa-1/genética , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Reparación del ADN por Recombinación/efectos de los fármacos , Sarcoma/genética
13.
Biomolecules ; 10(9)2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32825059

RESUMEN

Rheumatoid arthritis (RA) is a systemic disease that leads to joint destruction. During the last decade, the therapy of RA has been principally based on biological drugs. Although the efficacy of biological therapy has been established, patients demonstrated a high heterogeneity in clinical response to treatment. Several genetic polymorphisms play a part in the different response to biological drugs. This review summarizes the pharmacogenetics of biological agents approved for clinical RA treatment. We reviewed PubMed papers published over the past 20 years (2000-2020), inserting as the search term "rheumatoid arthritis and polymorphisms". Despite some studies showing important correlations between genetic polymorphisms and response to biological therapy in RA patients, most of these findings are still lacking and inconsistent. The personalized treatment according to a pharmacogenetics approach is promising but the available pharmacogenetics data on biological treatment in RA are not adequate and reliable to recommend pharmacogenetic tests before starting biological therapy in RA patients.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Factores Biológicos/farmacología , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Artritis Reumatoide/genética , Humanos , Polimorfismo de Nucleótido Simple/genética
14.
Nat Commun ; 11(1): 2393, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32409712

RESUMEN

Despite high initial efficacy, targeted therapies eventually fail in advanced cancers, as tumors develop resistance and relapse. In contrast to the substantial body of research on the molecular mechanisms of resistance, understanding of how resistance evolves remains limited. Using an experimental model of ALK positive NSCLC, we explored the evolution of resistance to different clinical ALK inhibitors. We found that resistance can originate from heterogeneous, weakly resistant subpopulations with variable sensitivity to different ALK inhibitors. Instead of the commonly assumed stochastic single hit (epi) mutational transition, or drug-induced reprogramming, we found evidence for a hybrid scenario involving the gradual, multifactorial adaptation to the inhibitors through acquisition of multiple cooperating genetic and epigenetic adaptive changes. Additionally, we found that during this adaptation tumor cells might present unique, temporally restricted collateral sensitivities, absent in therapy naïve or fully resistant cells, suggesting the potential for new therapeutic interventions, directed against evolving resistance.


Asunto(s)
Quinasa de Linfoma Anaplásico/antagonistas & inhibidores , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Quinasa de Linfoma Anaplásico/genética , Animales , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Resistencia a Antineoplásicos/genética , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Lapatinib/farmacología , Lapatinib/uso terapéutico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Polimorfismo de Nucleótido Simple/efectos de los fármacos , RNA-Seq , Análisis de la Célula Individual , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Psychopharmacology (Berl) ; 237(7): 2151-2159, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32382784

RESUMEN

INTRODUCTION: We explore the transcription factors involved in the molecular mechanism of antipsychotic (AP)-induced acute extrapyramidalsymptoms (EPS) in order to identify new candidate genes for pharmacogenetic studies. METHODS: Protein-protein interaction (PPI) networks previously created from three pharmacogenomic models (in vitro, animal, and peripheral blood inhumans) were used to, by means of several bioinformatic tools; identify key transcription factors (TFs) that regulate each network. Once the TFs wereidentified, SNPs disrupting the binding sites (TFBS) of these TFs in the genes of each network were selected for genotyping. Finally, SNP-basedassociations with EPS were analyzed in a sample of 356 psychiatric patients receiving AP. RESULTS: Our analysis identified 33 TFs expressed in the striatum, and 125 SNPs disrupting TFBS in 50 genes of our initial networks. Two SNPs (rs938112,rs2987902) in two genes (LSMAP and ABL1) were significantly associated with AP induced EPS (p < 0.001). These SNPs disrupt TFBS regulated byPOU2F1. CONCLUSION: Our results highlight the possible role of the disruption of TFBS by SNPs in the pharmacological response to AP.


Asunto(s)
Antipsicóticos/efectos adversos , Enfermedades de los Ganglios Basales/inducido químicamente , Enfermedades de los Ganglios Basales/genética , Farmacogenética/métodos , Pruebas de Farmacogenómica/métodos , Factores de Transcripción/genética , Animales , Enfermedades de los Ganglios Basales/metabolismo , Biología Computacional/métodos , Estudios de Seguimiento , Humanos , Estudios Longitudinales , Ratones , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Polimorfismo de Nucleótido Simple/fisiología , Estudios Prospectivos , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Factores de Transcripción/biosíntesis
16.
Pharmacogenomics ; 21(8): 509-520, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32427048

RESUMEN

Aim: GDF15 levels are a biomarker for metformin use. We performed the functional annotation of noncoding genome-wide association study (GWAS) SNPs for GDF15 levels and the Genotype-Tissue Expression (GTEx)-expression quantitative trait loci (eQTLs) for GDF15 expression within metformin-activated enhancers around GDF15. Materials & methods: These enhancers were identified using chromatin immunoprecipitation followed by sequencing data for active (H3K27ac) and silenced (H3K27me3) histone marks on human hepatocytes treated with metformin, Encyclopedia of DNA Elements data and cis-regulatory elements assignment tools. Results: The GWAS lead SNP rs888663, the SNP rs62122429 associated with GDF15 levels in the Outcome Reduction with Initial Glargine Intervention trial, and the GTEx-expression quantitative trait locus rs4808791 for GDF15 expression in whole blood are located in a metformin-activated enhancer upstream of GDF15 and tightly linked in Europeans and East Asians. Conclusion: Noncoding variation within a metformin-activated enhancer may increase GDF15 expression and help to predict GDF15 levels.


Asunto(s)
Estudio de Asociación del Genoma Completo/métodos , Factor 15 de Diferenciación de Crecimiento/biosíntesis , Factor 15 de Diferenciación de Crecimiento/genética , Metformina/farmacología , Polimorfismo de Nucleótido Simple/genética , Línea Celular , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Hipoglucemiantes/farmacología , Polimorfismo de Nucleótido Simple/efectos de los fármacos
17.
Ann Diagn Pathol ; 46: 151482, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32145682

RESUMEN

Osteosarcoma (OS) is a class of cancer originating from the bone, affecting mainly children and young adults. Our previous study showed that MAPK7 gene overexpression was significantly associated with tumor progression, poor treatment response, and worse overall survival, suggesting that MAPK7 could play an important role in OS tumorigenesis. We have investigated if MAPK7 overexpression was a result of any genomic changes in OS tumor specimens. We identified five SNPs (Single Nucleotide Polymorphism) previously described in databases, dbSNP and COSMIC, and identified two single nucleotide substitution not yet described. We found, in prechemotherapy specimens, a significant association of MAPK7 rs2233072G allele variant with metastasis at diagnosis and relapse (0.0909 and 0.0455, respectively). In post-chemotherapy, rs1054206GG specimen's genotype was associated with osteoblastic histological type (P= 0.0249) and presented decreased MAPK7 gene expression when compared with pre-chemotherapy specimens of same patients (P = 0.0095). Interestingly, it was observed some SNPs genotype exchange after chemotherapy. Our data indicated that MAPK7 gene expression associated with genotype exchange after chemotherapy, and these SNPs associated with important clinical parameters might be a valuable indicator for predicting in OS.


Asunto(s)
Neoplasias Óseas/genética , Predisposición Genética a la Enfermedad/genética , Proteína Quinasa 7 Activada por Mitógenos/genética , Osteosarcoma/genética , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Neoplasias Óseas/tratamiento farmacológico , Genotipo , Humanos , Osteosarcoma/tratamiento farmacológico , Pronóstico
18.
Adipocyte ; 9(1): 77-86, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32008426

RESUMEN

Whether the Adrenoceptor Beta 3 (ADRB3) gene rs4994 polymorphism could affect the individual risk of childhood and adolescent overweight/obesity remains controversial. This meta-analysis was performed to estimate the prevalence of this polymorphism in overweight/obesity, and test the potential association by summarizing existing evidence. Comprehensive literature search in PubMed, Web of Science, Cochrane Library, Wanfang, and CNKI databases was performed to identify eligible data sets. Finally, 16 studies involving 5,147 overweight/obese cases and 7,350 non-obese controls were included for further synthetic analyses. Odds ratio (OR) and its corresponding 95% confidence intervals (CIs) were statistically calculated. Totally, 69.9% of the included subjects came from East Asia. In the meta-analysis for overall population, statistically significant associations with increased risk of childhood and adolescent overweight/obesity were identified in allele model (OR 1.23, 95% CI 1.10-1.38), heterozygote model (OR 1.39, 95% CI 1.16-1.68), and dominant model (OR 1.31, 95% CI 1.12-1.54). Further stratified analysis according to geographical regions revealed that the statistical significance could only be detected in the East Asia subgroup in allele model, homozygote model, heterozygote model, and dominant model. In summary, our meta-analysis indicated that the ADRB3 rs4994 polymorphism could significantly increase the risk of childhood and adolescent overweight/obesity, especially for the East Asia's population.


Asunto(s)
Obesidad/genética , Sobrepeso/genética , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Receptores Adrenérgicos beta 3/genética , Adolescente , Niño , Asia Oriental , Genotipo , Humanos , Factores de Riesgo
19.
Pharmacogenomics J ; 20(3): 415-425, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31792371

RESUMEN

The aim of the study was to validate the impact of the single-nucleotide polymorphism rs2413739 (T > C) in the PACSIN2 gene on thiopurines pharmacological parameters and clinical response in an Italian cohort of pediatric patients with acute lymphoblastic leukemia (ALL) and inflammatory bowel disease (IBD). In ALL, PACSIN2 rs2413739 T allele was associated with a significant reduction of TPMT activity in erythrocytes (p = 0.0094, linear mixed-effect model, multivariate analysis considering TPMT genotype) and increased severe gastrointestinal toxicity during consolidation therapy (p = 0.049). A similar trend was present also for severe hematological toxicity during maintenance. In IBD, no significant effect of rs2413739 could be found on TPMT activity, however azathioprine effectiveness was reduced in patients carrying the T allele (linear mixed effect, p = 0.0058). In PBMC from healthy donors, a positive correlation between PACSIN2 and TPMT protein concentration could be detected (linear mixed effect, p = 0.045). These results support the role of PACSIN2 polymorphism on TPMT activity and mercaptopurine adverse effects in patients with ALL. Further evidence on PBMC and pediatric patients with IBD supports an association between PACSIN2 variants, TPMT activity, and thiopurines effects, even if more studies are needed since some of these effects may be tissue specific.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Azatioprina/farmacocinética , Enfermedades Inflamatorias del Intestino/genética , Polimorfismo de Nucleótido Simple/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adolescente , Adulto , Azatioprina/efectos adversos , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/metabolismo , Italia/epidemiología , Masculino , Mercaptopurina/efectos adversos , Mercaptopurina/farmacocinética , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo
20.
Addict Biol ; 25(5): e12806, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-31267641

RESUMEN

Opioid use among pregnant women is a growing public health concern in the United States. Infants exposed to opioids in utero are at risk of exhibiting neonatal opioid withdrawal syndrome (NOWS). The biological mechanisms underlying short and long-term consequences of in utero opioid exposure and NOWS are unknown. A potential genetic factor is a single-nucleotide polymorphism (SNP) in the mu-opioid receptor gene (OPRM1 A118G). Opioid exposed infants with the G-allele spend less time in hospitals after birth. To determine whether this SNP modulates the neurobehavioral effects of neonatal opioid exposure and withdrawal, we used mice possessing the equivalent Oprm1 SNP (A112G). Pups were treated chronically with saline or morphine from postnatal days (PNDs) 1 to 14, a developmental period equivalent to the third trimester of a human pregnancy and a sensitive period for opioid exposure in rodents. Morphine treatment produced significant developmental delays regardless of genotype and increased total ultrasonic vocalizations in males during spontaneous withdrawal. Animals were aged and tested for anxiety and drug response during adolescence and adulthood, respectively. AA morphine-treated animals showed reduced activity in the marble burying task compared with saline controls; however, this effect was absent in AG and GG animals. As adults, AA males exposed to morphine from PNDs 1 to 14 exhibited enhanced development of locomotor sensitization to morphine, whereas females showed reduced locomotor sensitization. These data suggest the involvement of the Oprm1 SNP for certain outcomes of neonatal opioid exposure and highlight the importance of considering sex and genetic variability for the prognosis of NOWS.


Asunto(s)
Analgésicos Opioides/farmacología , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Trastornos Relacionados con Opioides/genética , Polimorfismo de Nucleótido Simple/genética , Efectos Tardíos de la Exposición Prenatal/genética , Receptores Opioides mu/genética , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Embarazo , Receptores Opioides mu/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA