Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Blood ; 143(11): 1032-1044, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38096369

RESUMEN

ABSTRACT: Extreme disease phenotypes can provide key insights into the pathophysiology of common conditions, but studying such cases is challenging due to their rarity and the limited statistical power of existing methods. Herein, we used a novel approach to pathway-based mutational burden testing, the rare variant trend test (RVTT), to investigate genetic risk factors for an extreme form of sepsis-induced coagulopathy, infectious purpura fulminans (PF). In addition to prospective patient sample collection, we electronically screened over 10.4 million medical records from 4 large hospital systems and identified historical cases of PF for which archived specimens were available to perform germline whole-exome sequencing. We found a significantly increased burden of low-frequency, putatively function-altering variants in the complement system in patients with PF compared with unselected patients with sepsis (P = .01). A multivariable logistic regression analysis found that the number of complement system variants per patient was independently associated with PF after controlling for age, sex, and disease acuity (P = .01). Functional characterization of PF-associated variants in the immunomodulatory complement receptors CR3 and CR4 revealed that they result in partial or complete loss of anti-inflammatory CR3 function and/or gain of proinflammatory CR4 function. Taken together, these findings suggest that inherited defects in CR3 and CR4 predispose to the maladaptive hyperinflammation that characterizes severe sepsis with coagulopathy.


Asunto(s)
Púrpura Fulminante , Sepsis , Humanos , Púrpura Fulminante/genética , Estudios Prospectivos , Receptores de Complemento
2.
iScience ; 26(9): 107641, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37670779

RESUMEN

Targeted genome editing holds great promise in biology. However, efficient genome modification, including gene knock-in (KI), remains an unattained goal in multiple cell types and loci due to poor transfection efficiencies and low target genes expression, impeding the positive selection of recombined cells. Here, we describe a genome editing approach to achieve efficient gene targeting using hard to transfect erythroid cell lines. We demonstrate robust fluorescent protein KI efficiency in low expressed transcription factor (TF) genes (e.g., Myb or Zeb1). We further show the ability to target two independent loci in individual cells, exemplified by MYB-GFP and NuMA-Cherry double KI, allowing multicolor labeling of regulatory factors at physiological endogenous levels. Our KI tagging approach allowed us to perform genome-wide TF analysis at increased signal-to-noise ratios, and highlighted previously unidentified MYB target genes and pathways. Overall, we establish a versatile CRISPR-Cas9-based platform, offering attractive opportunities for the dissection of the erythroid differentiation process.

3.
Cancer Discov ; 13(6): 1478-1497, 2023 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-36847506

RESUMEN

Oncogenic mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 occur in a wide range of cancers, including acute myeloid leukemia (AML) and glioma. Mutant IDH enzymes convert 2-oxoglutarate (2OG) to (R)-2-hydroxyglutarate [(R)-2HG], an oncometabolite that is hypothesized to promote cellular transformation by dysregulating 2OG-dependent enzymes. The only (R)-2HG target that has been convincingly shown to contribute to transformation by mutant IDH is the myeloid tumor suppressor TET2. However, there is ample evidence to suggest that (R)-2HG has other functionally relevant targets in IDH-mutant cancers. Here, we show that (R)-2HG inhibits KDM5 histone lysine demethylases and that this inhibition contributes to cellular transformation in IDH-mutant AML and IDH-mutant glioma. These studies provide the first evidence of a functional link between dysregulation of histone lysine methylation and transformation in IDH-mutant cancers. SIGNIFICANCE: Mutant IDH is known to induce histone hypermethylation. However, it is not known if this hypermethylation is functionally significant or is a bystander effect of (R)-2HG accumulation in IDH-mutant cells. Here, we provide evidence that KDM5 inhibition by (R)-2HG contributes to mutant IDH-mediated transformation in AML and glioma. This article is highlighted in the In This Issue feature, p. 1275.


Asunto(s)
Glioma , Leucemia Mieloide Aguda , Humanos , Histonas/metabolismo , Histona Demetilasas/genética , Mutación , Glutaratos , Transformación Celular Neoplásica/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Glioma/genética , Metilación de ADN , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo
4.
NPJ Precis Oncol ; 6(1): 61, 2022 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-36056177

RESUMEN

The mutant IDH1 inhibitor ivosidenib improves outcomes for patients with IDH1-mutated cholangiocarcinoma, but resistance inevitably develops. Mechanisms of resistance and strategies to overcome resistance are poorly understood. Here we describe two patients with IDH1 R132C-mutated metastatic cholangiocarcinoma who developed acquired resistance to ivosidenib. After disease progression, one patient developed an oncogenic IDH2 mutation, and the second patient acquired a secondary IDH1 D279N mutation. To characterize the putative IDH1 resistance mutation, cells expressing the double-mutant were generated. In vitro, IDH1 R132H/D279N produces (R)-2HG less efficiently than IDH1 R132H. However, its binding to ivosidenib is impaired and it retains the ability to produce (R)-2HG and promote cellular transformation in the presence of ivosidenib. The irreversible mutant IDH1 inhibitor LY3410738 binds and blocks (R)-2HG production and cellular transformation by IDH1 R132H/D279N. These resistance mechanisms suggest that IDH1-mutated cholangiocarcinomas remain dependent on (R)-2HG even after prolonged ivosidenib treatment. Sequential mutant IDH inhibitor therapy should be explored as a strategy to overcome acquired resistance to mutant IDH inhibitors.

5.
Sci Rep ; 8(1): 16875, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30443025

RESUMEN

Besides their well-known roles in digestion and fat solubilization, bile acids (BAs) have been described as signaling molecules activating the nuclear receptor Farnesoid-X-receptor (FXRα) or the G-protein-coupled bile acid receptor-1 (GPBAR-1 or TGR5). In previous reports, we showed that BAs decrease male fertility due to abnormalities of the germ cell lineage dependent on Tgr5 signaling pathways. In the presentstudy, we tested whether BA exposure could impact germ cell DNA integrity leading to potential implications for progeny. For that purpose, adult F0 male mice were fed a diet supplemented with cholic acid (CA) or the corresponding control diet during 3.5 months prior mating. F1 progeny from CA exposed founders showed higher perinatal lethality, impaired BA homeostasis and reduced postnatal growth, as well as altered glucose metabolism in later life. The majority of these phenotypic traits were maintained up to the F2 generation. In F0 sperm cells, differential DNA methylation associated with CA exposure may contribute to the initial programming of developmental and metabolic defects observed in F1 and F2 offspring. Tgr5 knock-out mice combined with in vitro strategies defined the critical role of paternal Tgr5 dependent pathways in the multigenerational impacts of ancestral CA exposure.


Asunto(s)
Bilis/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Animales , Linaje de la Célula/efectos de los fármacos , Ácido Cólico/farmacología , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Dieta , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Fenotipo , Transducción de Señal/efectos de los fármacos , Espermatozoides/citología , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo , ADN Metiltransferasa 3B
6.
Stem Cell Reports ; 11(4): 944-958, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30245210

RESUMEN

Several studies have reported an association between the farnesoid X receptor alpha (FXRα) and estrogenic signaling pathways. Fxrα could thus be involved in the reprotoxic effects of endocrine disruptors such as bisphenol-A (BPA). To test this hypothesis, mice were exposed to BPA and/or stigmasterol (S), an FXRα antagonist. Following the exposure to both molecules, wild-type animals showed impaired fertility and lower sperm cell production associated with the alteration of the establishment and maintenance of the undifferentiated germ cell pool. The crosstalk between BPA and FXRα is further supported by the lower impact of BPA in mice genetically ablated for Fxrα and the fact that BPA counteracted the effects of FXRα agonists. These effects might result from the downregulation of Fxrα expression following BPA exposure. BPA and S act additively in human testis. Our data demonstrate that FXRα activity modulates the impact of BPA on male gonads and on undifferentiated germ cell population.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Diferenciación Celular , Células Germinativas/patología , Homeostasis , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Fenoles/toxicidad , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal , Adulto , Animales , Animales Recién Nacidos , Diferenciación Celular/efectos de los fármacos , Feto/efectos de los fármacos , Feto/patología , Células Germinativas/efectos de los fármacos , Células Germinativas/metabolismo , Homeostasis/efectos de los fármacos , Humanos , Masculino , Ratones , Persona de Mediana Edad , Células de Sertoli/efectos de los fármacos , Células de Sertoli/metabolismo , Transducción de Señal/efectos de los fármacos , Estigmasterol/toxicidad
7.
Sci Rep ; 7: 42182, 2017 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-28181583

RESUMEN

Bile acids (BAs) are molecules with endocrine activities controlling several physiological functions such as immunity, glucose homeostasis, testicular physiology and male fertility. The role of the nuclear BA receptor FXRα in the control of BA homeostasis has been well characterized. The present study shows that testis synthetize BAs. We demonstrate that mice invalidated for the gene encoding FXRα have altered BA homeostasis in both liver and testis. In the absence of FXRα, BA exposure differently alters hepatic and testicular expression of genes involved in BA synthesis. Interestingly, Fxrα-/- males fed a diet supplemented with BAs show alterations of testicular physiology and sperm production. This phenotype was correlated with the altered testicular BA homeostasis and the production of intermediate metabolites of BAs which led to the modulation of CAR signaling pathways within the testis. The role of the CAR signaling pathways within testis was validated using specific CAR agonist (TCPOBOP) and inverse agonist (androstanol) that respectively inhibited or reproduced the phenotype observed in Fxrα-/- males fed BA-diet. These data open interesting perspectives to better define how BA homeostasis contributes to physiological or pathophysiological conditions via the modulation of CAR activity.


Asunto(s)
Ácidos y Sales Biliares/genética , Receptores Citoplasmáticos y Nucleares/genética , Reproducción/genética , Testículo/metabolismo , Androstanoles/farmacología , Animales , Ácidos y Sales Biliares/biosíntesis , Receptor de Androstano Constitutivo , Regulación de la Expresión Génica/efectos de los fármacos , Homeostasis/genética , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Espermatozoides/metabolismo
8.
Stem Cell Reports ; 9(1): 315-328, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28669602

RESUMEN

Spermatogenesis is the process by which spermatozoa are generated from spermatogonia. This cell population is heterogeneous, with self-renewing spermatogonial stem cells (SSCs) and progenitor spermatogonia that will continue on a path of differentiation. Only SSCs have the ability to regenerate and sustain spermatogenesis. This makes the testis a good model to investigate stem cell biology. The Farnesoid X Receptor alpha (FXRα) was recently shown to be expressed in the testis. However, its global impact on germ cell homeostasis has not yet been studied. Here, using a phenotyping approach in Fxrα-/- mice, we describe unexpected roles of FXRα on germ cell physiology independent of its effects on somatic cells. FXRα helps establish and maintain an undifferentiated germ cell pool and in turn influences male fertility. FXRα regulates the expression of several pluripotency factors. Among these, in vitro approaches show that FXRα controls the expression of the pluripotency marker Lin28 in the germ cells.


Asunto(s)
Receptores Citoplasmáticos y Nucleares/metabolismo , Espermatogénesis , Espermatozoides/citología , Envejecimiento , Animales , Células Cultivadas , Femenino , Fertilidad , Eliminación de Gen , Regulación de la Expresión Génica , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones Endogámicos C57BL , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Reproducción , Células de Sertoli/citología , Células de Sertoli/metabolismo , Espermatozoides/metabolismo , Testículo/citología , Testículo/metabolismo , Testículo/ultraestructura
9.
Oncotarget ; 7(15): 19468-82, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-26848619

RESUMEN

The bile acid receptor Farnesol-X-Receptor alpha (FRXα) is a member of the nuclear receptor superfamily. FRXα is expressed in the interstitial compartment of the adult testes, which contain the Leydig cells. In adult, short term treatment (12 hours) with FRXα agonist inhibits the expression of steroidogenic genes via the induction of the Small heterodimer partner (SHP). However the consequences of FRXα activation on testicular pathophysiology have never been evaluated. We demonstrate here that mice fed a diet supplemented with bile acid during pubertal age show increased incidence of infertility. This is associated with altered differentiation and increase apoptosis of germ cells due to lower testosterone levels. At the molecular level, next to the repression of basal steroidogenesis via the induction expression of Shp and Dax-1, two repressors of steroidogenesis, the main action of the BA-FRXα signaling is through lowering the Leydig cell sensitivity to the hypothalamo-pituitary axis, the main regulator of testicular endocrine function. In conclusion, BA-FRXα signaling is a critical actor during sexual maturation.


Asunto(s)
Ácidos y Sales Biliares/farmacología , Receptores Citoplasmáticos y Nucleares/metabolismo , Maduración Sexual/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Ácidos y Sales Biliares/administración & dosificación , Línea Celular , Ácido Cólico/administración & dosificación , Ácido Cólico/farmacología , Fertilidad/efectos de los fármacos , Isoxazoles/farmacología , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Interferencia de ARN , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal/genética , Testículo/citología , Testículo/metabolismo , Testosterona/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA