Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Cell Commun Signal ; 21(1): 328, 2023 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-37974198

RESUMEN

BACKGROUND: Glioblastoma is the most common and aggressive primary brain tumor with extremely poor prognosis, highlighting an urgent need for developing novel treatment options. Identifying epigenetic vulnerabilities of cancer cells can provide excellent therapeutic intervention points for various types of cancers. METHOD: In this study, we investigated epigenetic regulators of glioblastoma cell survival through CRISPR/Cas9 based genetic ablation screens using a customized sgRNA library EpiDoKOL, which targets critical functional domains of chromatin modifiers. RESULTS: Screens conducted in multiple cell lines revealed ASH2L, a histone lysine methyltransferase complex subunit, as a major regulator of glioblastoma cell viability. ASH2L depletion led to cell cycle arrest and apoptosis. RNA sequencing and greenCUT&RUN together identified a set of cell cycle regulatory genes, such as TRA2B, BARD1, KIF20B, ARID4A and SMARCC1 that were downregulated upon ASH2L depletion. Mass spectrometry analysis revealed the interaction partners of ASH2L in glioblastoma cell lines as SET1/MLL family members including SETD1A, SETD1B, MLL1 and MLL2. We further showed that glioblastoma cells had a differential dependency on expression of SET1/MLL family members for survival. The growth of ASH2L-depleted glioblastoma cells was markedly slower than controls in orthotopic in vivo models. TCGA analysis showed high ASH2L expression in glioblastoma compared to low grade gliomas and immunohistochemical analysis revealed significant ASH2L expression in glioblastoma tissues, attesting to its clinical relevance. Therefore, high throughput, robust and affordable screens with focused libraries, such as EpiDoKOL, holds great promise to enable rapid discovery of novel epigenetic regulators of cancer cell survival, such as ASH2L. CONCLUSION: Together, we suggest that targeting ASH2L could serve as a new therapeutic opportunity for glioblastoma. Video Abstract.


Asunto(s)
Glioblastoma , Proteínas Nucleares , Humanos , Supervivencia Celular , Proteínas Nucleares/metabolismo , Glioblastoma/genética , Sistemas CRISPR-Cas/genética , ARN Guía de Sistemas CRISPR-Cas , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Cinesinas/genética , Cinesinas/metabolismo
2.
J Cardiothorac Surg ; 18(1): 189, 2023 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-37312199

RESUMEN

BACKGROUND: Lung cancer is known as the most common and highly metastatic form of cancer worldwide. Tumour node metastasis (TNM) staging is the gold standard classification system for the decision-making process for appropriate treatment. Particularly N status has the most important prognostic value in the absence of distant metastasis. Traditional diagnostic methods are capable of detecting metastasis; however, they may fail to detect micrometastasis, which plays a role in disease recurrence and patients' long-term survival. Occult micrometastasis can change the tumour's TNM staging and, consequently, the patient's treatment regimen. METHODS: The median number of three lymph node tissues were collected from 30 patients who underwent surgery for non-small cell lung cancer. Lymph node tissues were collected from different lymph node stations according to the location of the patient's tumour. CK19, EpCAM and CEACAM5 gene expressions were analysed in tissues using quantitative real-time polymerase chain reaction to detect micrometastasis in distant lymph nodes. RESULTS: Triple positivity was seen in 26 out of 30 patients which 19 patients were upstaged from N0 to N2. While survival was not significantly affected between upstaged and non-upstaged patients, patients upstaged with multiple-station N2 had a significantly higher recurrence and lower survival compared to single-station N2. CONCLUSION: A combination of CK19, EpCAM and CEACAM5 gene expressions in lymph nodes can be used to identify micrometastasis which postoperatively may be used as a tool to predict patients' recurrence and survival.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Antígeno Carcinoembrionario , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/cirugía , Molécula de Adhesión Celular Epitelial/genética , Expresión Génica , Proteínas Ligadas a GPI , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/cirugía , Ganglios Linfáticos , Micrometástasis de Neoplasia/genética , Pronóstico
3.
Cell Death Dis ; 13(8): 710, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35973998

RESUMEN

Dysregulation of the epigenome due to alterations in chromatin modifier proteins commonly contribute to malignant transformation. To interrogate the roles of epigenetic modifiers in cancer cells, we generated an epigenome-wide CRISPR-Cas9 knockout library (EPIKOL) that targets a wide-range of epigenetic modifiers and their cofactors. We conducted eight screens in two different cancer types and showed that EPIKOL performs with high efficiency in terms of sgRNA distribution and depletion of essential genes. We discovered novel epigenetic modifiers that regulate triple-negative breast cancer (TNBC) and prostate cancer cell fitness. We confirmed the growth-regulatory functions of individual candidates, including SS18L2 and members of the NSL complex (KANSL2, KANSL3, KAT8) in TNBC cells. Overall, we show that EPIKOL, a focused sgRNA library targeting ~800 genes, can reveal epigenetic modifiers that are essential for cancer cell fitness under in vitro and in vivo conditions and enable the identification of novel anti-cancer targets. Due to its comprehensive epigenome-wide targets and relatively high number of sgRNAs per gene, EPIKOL will facilitate studies examining functional roles of epigenetic modifiers in a wide range of contexts, such as screens in primary cells, patient-derived xenografts as well as in vivo models.


Asunto(s)
Sistemas CRISPR-Cas , Neoplasias de la Mama Triple Negativas , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Cromatina , Detección Precoz del Cáncer , Humanos , Masculino , Neoplasias de la Mama Triple Negativas/genética
4.
Nat Cell Biol ; 24(4): 554-564, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35411083

RESUMEN

Epithelial-mesenchymal transition (EMT) programs operate within carcinoma cells, where they generate phenotypes associated with malignant progression. In their various manifestations, EMT programs enable epithelial cells to enter into a series of intermediate states arrayed along the E-M phenotypic spectrum. At present, we lack a coherent understanding of how carcinoma cells control their entrance into and continued residence in these various states, and which of these states favour the process of metastasis. Here we characterize a layer of EMT-regulating machinery that governs E-M plasticity (EMP). This machinery consists of two chromatin-modifying complexes, PRC2 and KMT2D-COMPASS, which operate as critical regulators to maintain a stable epithelial state. Interestingly, loss of these two complexes unlocks two distinct EMT trajectories. Dysfunction of PRC2, but not KMT2D-COMPASS, yields a quasi-mesenchymal state that is associated with highly metastatic capabilities and poor survival of patients with breast cancer, suggesting that great caution should be applied when PRC2 inhibitors are evaluated clinically in certain patient cohorts. These observations identify epigenetic factors that regulate EMP, determine specific intermediate EMT states and, as a direct consequence, govern the metastatic ability of carcinoma cells.


Asunto(s)
Neoplasias de la Mama , Carcinoma , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/genética , Femenino , Humanos , Metástasis de la Neoplasia/patología
5.
Stem Cell Res ; 54: 102438, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34214898

RESUMEN

Friedreich's ataxia (FRDA) is a rare neurodegenerative disorder which is caused by triplet repeat expansion (GAA) in the first intron of FXN gene. In this present study, we generated induced pluripotent stem cells (iPSC) lines from fibroblasts of three unrelated FRDA patients using integration-free episomal vectors. All iPSC lines express the pluripotency markers such as OCT4 and SSEA4, display normal karyotypes and can differentiate into all three germ layers via in vivo teratoma formation assay.


Asunto(s)
Ataxia de Friedreich , Células Madre Pluripotentes Inducidas , Proteínas de Unión a Hierro , Ataxia de Friedreich/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Intrones/genética , Proteínas de Unión a Hierro/genética , Proteínas de Unión a Hierro/metabolismo , Expansión de Repetición de Trinucleótido , Frataxina
6.
iScience ; 24(5): 102425, 2021 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-34036247

RESUMEN

Elucidations of the factors that promote the growth of disseminated tumor cells (DTCs) into life-threatening lesions stand to provide much needed prognostic and therapeutic targets of translational utility for patients with metastatic cancer. To identify such regulators, we conducted gain-of-function cDNA library screening to discover genes that foster prostate cancer cell colonization of mouse lungs as an experimental model. Our efforts identified the metabolic enzyme aldolase A (ALDOA) as a driver of cancer cell motility, anchorage-independent growth, and metastatic colonization, and as a prognosticator of adverse patient outcome across many malignancies, including prostate, breast, pancreatic, and liver cancers. Metabolomics coupled with biochemical and functional analyses revealed that ALDOA triggered the activation of adenosine-5'-monophosphate (AMP)-activated protein kinase (AMPK), which we demonstrate played essential promalignant activities in ALDOA-expressing cells. Collectively, these findings unveiled vivo approaches to identify metastatic colonization regulators and uncovered previously undescribed roles for ALDOA-AMPK pathway in tumor progression.

7.
Stem Cell Res Ther ; 12(1): 287, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33985578

RESUMEN

BACKGROUND: Griscelli syndrome type 2 (GS-2) is a rare, autosomal recessive immune deficiency syndrome caused by a mutation in the RAB27A gene, which results in the absence of a protein involved in vesicle trafficking and consequent loss of function of in particular cytotoxic T and NK cells. Induced pluripotent stem cells (iPSC) express genes associated with pluripotency, have the capacity for infinite expansion, and can differentiate into cells from all three germ layers. They can be induced using integrative or non-integrative systems for transfer of the Oct4, Sox2, Klf4, and cMyc (OSKM) transcription factors. To better understand the pathophysiology of GS-2 and to test novel treatment options, there is a need for an in vitro model of GS-2. METHODS: Here, we generated iPSCs from 3 different GS-2 patients using lentiviral vectors. The iPSCs were characterized using flow cytometry and RT-PCR and tested for the expression of pluripotency markers. In vivo differentiation to cells from all three germlines was tested using a teratoma assay. In vitro differentiation of GS-2 iPSCs into hematopoietic stem and progenitor cells was done using Op9 feeder layers and specified media. RESULTS: All GS-2 iPSC clones displayed a normal karyotype (46XX or 46XY) and were shown to express the same RAB27A gene mutation that was present in the original somatic donor cells. GS-2 iPSCs expressed SSEA1, SSEA4, TRA-1-60, TRA-1-81, and OCT4 proteins, and SOX2, NANOG, and OCT4 expression were confirmed by RT-PCR. Differentiation capacity into cells from all three germ layers was confirmed using the teratoma assay. GS-2 iPSCs showed the capacity to differentiate into cells of the hematopoietic lineage. CONCLUSIONS: Using the lentiviral transfer of OSKM, we were able to generate different iPSC clones from 3 GS-2 patients. These cells can be used in future studies for the development of novel treatment options and to study the pathophysiology of GS-2 disease.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Pluripotentes Inducidas , Diferenciación Celular , Células Nutrientes , Humanos , Factor 4 Similar a Kruppel , Linfohistiocitosis Hemofagocítica , Piebaldismo , Enfermedades de Inmunodeficiencia Primaria
8.
Cell Death Dis ; 11(8): 658, 2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32814763

RESUMEN

Complete hydatidiform mole (HM) is a gestational trophoblastic disease resulting in hyperproliferation of trophoblast cells and absence of embryo development. Mutations in the maternal-effect gene NLRP7 are the major cause of familial recurrent complete HM. Here, we established an in vitro model of HM using patient-specific induced pluripotent stem cells (iPSCs) derived trophoblasts harboring NLRP7 mutations. Using whole transcriptome profiling during trophoblast differentiation, we showed that impaired NLRP7 expression results in precocious downregulation of pluripotency factors, activation of trophoblast lineage markers, and promotes maturation of differentiated extraembryonic cell types such as syncytiotrophoblasts. Interestingly, we found that these phenotypes are dependent on BMP4 signaling and BMP pathway inhibition corrected the excessive trophoblast differentiation of patient-derived iPSCs. Our human iPSC model of a genetic placental disease recapitulates aspects of trophoblast biology, highlights the broad utility of iPSC-derived trophoblasts for modeling human placental diseases and identifies NLRP7 as an essential modulator of key developmental cell fate regulators.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Trofoblastos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteína Morfogenética Ósea 4/fisiología , Diferenciación Celular/genética , Células Cultivadas , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Mola Hidatiforme/genética , Mola Hidatiforme/fisiopatología , Células Madre Pluripotentes Inducidas/fisiología , Modelos Biológicos , Placenta/metabolismo , Embarazo , Transducción de Señal/fisiología , Transcriptoma/genética
9.
Biol Reprod ; 102(6): 1270-1280, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32163131

RESUMEN

Human chorionic gonadotropin (hCG) is a luteotropic hormone that promotes the survival and steroidogenic activity of corpus luteum (CL) by acting through luteinizing hormone receptors (LHRs) expressed on luteinized theca and granulosa cells (GCs). Therefore, it is used to support luteal phase in in vitro fertilization (IVF) cycles to improve clinical pregnancy rates and prevent miscarriage. However, the molecular mechanism underlying this action of hCG is not well characterized. To address this question, we designed an in vitro translational research study on the luteal GCs obtained from 58 IVF patients. hCG treatment at different concentrations and time points activated c-Jun N-terminal kinase (JNK) pathway and significantly increased its endogenous kinase activity along with upregulated expression of steroidogenic enzymes (steroidogenic acute regulatory protein (stAR), 3ß-Hydroxysteroid dehydrogenase (3ß-HSD)) in a dose-dependent manner in the luteal GCs. As a result, in vitro P production of the cells was significantly enhanced after hCG. When JNK pathway was inhibited pharmacologically or knocked-down with small interfering RNA luteal function was compromised, P4 production was declined along with the expression of stAR and 3ß-HSD in the cells. Further, hCG treatment after JNK inhibition failed to correct the luteal defect and promote P4 output. Similar to hCG, luteinizing hormone (LH) treatment improved luteal function as well and this action of LH was associated with JNK activation in the luteal GCs. These findings could be important from the perspective of CL biology and luteal phase in human because we for the first time identify a critical role for JNK signaling pathway downstream LHR activation by hCG/LH in luteal GCs. SUMMARY SENTENCE: JNK signaling pathway plays a central role in the upregulated expression of the steroidogenic enzymes StAR and 3b-HSD and augmented progesterone production by hCG/LH in human luteal granulosa cells.


Asunto(s)
Gonadotropina Coriónica/farmacología , Cuerpo Lúteo/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Progesterona/metabolismo , Adulto , Femenino , Fertilización In Vitro , Células de la Granulosa/metabolismo , Humanos , Hormona Luteinizante/farmacología
10.
Oncogene ; 39(10): 2187-2201, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31822799

RESUMEN

Androgen deprivation therapy (ADT) is the standard care for prostate cancer (PCa) patients who fail surgery or radiotherapy. While initially effective, the cancer almost always recurs as a more aggressive castration resistant prostate cancer (CRPC). Previous studies have demonstrated that chromatin modifying enzymes can play a critical role in the conversion to CRPC. However, only a handful of these potential pharmacological targets have been tested. Therefore, in this study, we conducted a focused shRNA screen of chromatin modifying enzymes previously shown to be involved in cellular differentiation. We found that altering the balance between histone methylation and demethylation impacted growth and proliferation. Of all genes tested, KDM3B, a histone H3K9 demethylase, was found to have the most antiproliferative effect. These results were phenocopied with a KDM3B CRISPR/Cas9 knockout. When tested in several PCa cell lines, the decrease in proliferation was remarkably specific to androgen-independent cells. Genetic rescue experiments showed that only the enzymatically active KDM3B could recover the phenotype. Surprisingly, despite the decreased proliferation of androgen-independent cell no alterations in the cell cycle distribution were observed following KDM3B knockdown. Whole transcriptome analyses revealed changes in the gene expression profile following loss of KDM3B, including downregulation of metabolic enzymes such as ARG2 and RDH11. Metabolomic analysis of KDM3B knockout showed a decrease in several critical amino acids. Overall, our work reveals, for the first time, the specificity and the dependence of KDM3B in CRPC proliferation.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Procesamiento Proteico-Postraduccional , Arginasa/genética , Línea Celular Tumoral , Perfilación de la Expresión Génica , Código de Histonas , Humanos , Masculino , Metilación , Oxidorreductasas/genética , Células PC-3 , Neoplasias de la Próstata Resistentes a la Castración/genética
11.
Stem Cell Reports ; 13(4): 627-641, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31522975

RESUMEN

Organoid technologies have become a powerful emerging tool to model liver diseases, for drug screening, and for personalized treatments. These applications are, however, limited in their capacity to generate functional hepatocytes in a reproducible and efficient manner. Here, we generated and characterized the hepatic organoid (eHEPO) culture system using human induced pluripotent stem cell (iPSC)-derived EpCAM-positive endodermal cells as an intermediate. eHEPOs can be produced within 2 weeks and expanded long term (>16 months) without any loss of differentiation capacity to mature hepatocytes. Starting from patient-specific iPSCs, we modeled citrullinemia type 1, a urea cycle disorder caused by mutations in the argininosuccinate synthetase (ASS1) enzyme. The disease-related ammonia accumulation phenotype in eHEPOs could be reversed by the overexpression of the wild-type ASS1 gene, which also indicated that this model is amenable to genetic manipulation. Thus, eHEPOs are excellent unlimited cell sources to generate functional hepatic organoids in a fast and efficient manner.


Asunto(s)
Diferenciación Celular , Susceptibilidad a Enfermedades , Endodermo/citología , Hepatocitos/citología , Hígado/citología , Hígado/embriología , Organogénesis , Organoides/citología , Biomarcadores , Molécula de Adhesión Celular Epitelial/genética , Molécula de Adhesión Celular Epitelial/metabolismo , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/citología , Técnicas de Cultivo de Tejidos
12.
Mol Cell Proteomics ; 18(9): 1756-1771, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31221721

RESUMEN

Epithelial-mesenchymal transition (EMT) is driven by complex signaling events that induce dramatic biochemical and morphological changes whereby epithelial cells are converted into cancer cells. However, the underlying molecular mechanisms remain elusive. Here, we used mass spectrometry based quantitative proteomics approach to systematically analyze the post-translational biochemical changes that drive differentiation of human mammary epithelial (HMLE) cells into mesenchymal. We identified 314 proteins out of more than 6,000 unique proteins and 871 phosphopeptides out of more than 7,000 unique phosphopeptides as differentially regulated. We found that phosphoproteome is more unstable and prone to changes during EMT compared with the proteome and multiple alterations at proteome level are not thoroughly represented by transcriptional data highlighting the necessity of proteome level analysis. We discovered cell state specific signaling pathways, such as Hippo, sphingolipid signaling, and unfolded protein response (UPR) by modeling the networks of regulated proteins and potential kinase-substrate groups. We identified two novel factors for EMT whose expression increased on EMT induction: DnaJ heat shock protein family (Hsp40) member B4 (DNAJB4) and cluster of differentiation 81 (CD81). Suppression of DNAJB4 or CD81 in mesenchymal breast cancer cells resulted in decreased cell migration in vitro and led to reduced primary tumor growth, extravasation, and lung metastasis in vivo Overall, we performed the global proteomic and phosphoproteomic analyses of EMT, identified and validated new mRNA and/or protein level modulators of EMT. This work also provides a unique platform and resource for future studies focusing on metastasis and drug resistance.


Asunto(s)
Neoplasias de la Mama/patología , Transición Epitelial-Mesenquimal/fisiología , Proteínas del Choque Térmico HSP40/metabolismo , Fosfoproteínas/metabolismo , Tetraspanina 28/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Movimiento Celular/fisiología , Transición Epitelial-Mesenquimal/genética , Femenino , Proteínas del Choque Térmico HSP40/genética , Humanos , Estimación de Kaplan-Meier , Neoplasias Mamarias Experimentales/patología , Ratones Desnudos , Reproducibilidad de los Resultados , Tetraspanina 28/genética
13.
In Vitro Cell Dev Biol Anim ; 55(7): 473-481, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31214928

RESUMEN

Leptin, a metabolic hormone, regulates the reproductive functions responding to both nutritional and body conditions. Embryonic stem cells play important roles in reproductive technology, but their derivation can be challenging. In this study, we evaluated the derivation rates of mouse embryonic stem cell (mESC) line from blastocysts developing in embryo culture media supplemented with different leptin concentrations. The results showed that addition of leptin into the embryo culture medium supported the in vitro development of mouse embryo. The mESC line derivation rates for media treated with 0, 10, 50, and 100 ng/ml of leptin were 61.24 % (54/88), 84.96 % (42/50), 81.79 % (61/76), and 85.78 % (56/67), respectively. In addition, leptin treatment of blastocysts upregulated the expression levels of the trophectoderm marker Cdx2, whereas inner cell mass markers Oct-4 and Nanog were not affected. mESC lines derived after leptin treatment demonstrated hallmarks of pluripotency, such as alkaline phosphatase activity, expression of, OCT4, NANOG, and SSEA1, as well as the ability to form embryoid bodies and well-differentiated teratomas. In conclusion, leptin has a positive effect on the derivation rate of mouse embryonic stem cell lines which may be, in part, due to its effects on the development of the trophectoderm cell lineage in the embryo.


Asunto(s)
Blastocisto/citología , Proliferación Celular/efectos de los fármacos , Leptina/farmacología , Células Madre Embrionarias de Ratones/citología , Teratoma/metabolismo , Animales , Factor de Transcripción CDX2/biosíntesis , Diferenciación Celular/efectos de los fármacos , Línea Celular , Linaje de la Célula , Medios de Cultivo/farmacología , Técnicas de Cultivo de Embriones , Cuerpos Embrioides/citología , Antígeno Lewis X/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteína Homeótica Nanog/biosíntesis , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Teratoma/inducido químicamente
14.
Stem Cell Reports ; 9(3): 943-955, 2017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28781076

RESUMEN

Organogenesis and tissue development occur through sequential stepwise processes leading to increased lineage restriction and loss of pluripotency. An exception to this appears in the adult human breast, where rare variant epithelial cells exhibit pluripotency and multilineage differentiation potential when removed from the signals of their native microenvironment. This phenomenon provides a unique opportunity to study mechanisms that lead to cellular reprogramming and lineage plasticity in real time. Here, we show that primary human mammary epithelial cells (HMECs) lose expression of differentiated mammary epithelial markers in a manner dependent on paracrine factors and epigenetic regulation. Furthermore, we demonstrate that HMEC reprogramming is dependent on gene silencing by the DNA methyltransferase DNMT3A and loss of histone transcriptional marks following downregulation of the methyltransferase DOT1L. These results demonstrate that lineage commitment in adult tissues is context dependent and highlight the plasticity of somatic cells when removed from their native tissue microenvironment.


Asunto(s)
Linaje de la Célula/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Epigénesis Genética , Células Epiteliales/metabolismo , Glándulas Mamarias Humanas/citología , Metiltransferasas/metabolismo , Diferenciación Celular/genética , Microambiente Celular , Reprogramación Celular/genética , ADN Metiltransferasa 3A , N-Metiltransferasa de Histona-Lisina , Histonas/metabolismo , Humanos , Metilación , Células del Estroma/citología
15.
Immunity ; 47(1): 183-198.e6, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28723550

RESUMEN

Tissue macrophages arise during embryogenesis from yolk-sac (YS) progenitors that give rise to primitive YS macrophages. Until recently, it has been impossible to isolate or derive sufficient numbers of YS-derived macrophages for further study, but data now suggest that induced pluripotent stem cells (iPSCs) can be driven to undergo a process reminiscent of YS-hematopoiesis in vitro. We asked whether iPSC-derived primitive macrophages (iMacs) can terminally differentiate into specialized macrophages with the help of growth factors and organ-specific cues. Co-culturing human or murine iMacs with iPSC-derived neurons promoted differentiation into microglia-like cells in vitro. Furthermore, murine iMacs differentiated in vivo into microglia after injection into the brain and into functional alveolar macrophages after engraftment in the lung. Finally, iPSCs from a patient with familial Mediterranean fever differentiated into iMacs with pro-inflammatory characteristics, mimicking the disease phenotype. Altogether, iMacs constitute a source of tissue-resident macrophage precursors that can be used for biological, pathophysiological, and therapeutic studies.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Hematopoyesis , Macrófagos/fisiología , Neuronas/fisiología , Células Madre Pluripotentes/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Embrión de Mamíferos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurogénesis
16.
Cell Death Dis ; 8(6): e2897, 2017 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-28661478

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can selectively kill tumor cells. TRAIL resistance in cancers is associated with aberrant expression of the key components of the apoptotic program. However, how these components are regulated at the epigenetic level is not understood. In this study, we investigated novel epigenetic mechanisms regulating TRAIL response in glioblastoma multiforme (GBM) cells by a short-hairpin RNA loss-of-function screen. We interrogated 48 genes in DNA and histone modification pathways and identified KDM2B, an H3K36-specific demethylase, as a novel regulator of TRAIL response. Accordingly, silencing of KDM2B significantly enhanced TRAIL sensitivity, the activation of caspase-8, -3 and -7 and PARP cleavage. KDM2B knockdown also accelerated the apoptosis, as revealed by live-cell imaging experiments. To decipher the downstream molecular pathways regulated by KDM2B, levels of apoptosis-related genes were examined by RNA-sequencing upon KDM2B loss, which revealed derepression of proapoptotic genes Harakiri (HRK), caspase-7 and death receptor 4 (DR4) and repression of antiapoptotic genes. The apoptosis phenotype was partly dependent on HRK upregulation, as HRK knockdown significantly abrogated the sensitization. KDM2B-silenced tumors exhibited slower growth in vivo. Taken together, our findings suggest a novel mechanism, where the key apoptosis components are under epigenetic control of KDM2B in GBM cells.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Proteínas F-Box/genética , Glioblastoma/genética , Histona Demetilasas con Dominio de Jumonji/genética , ARN Interferente Pequeño/genética , Apoptosis/genética , Caspasa 7/genética , Línea Celular Tumoral , Metilación de ADN/genética , Epigénesis Genética/genética , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Glioblastoma/patología , Código de Histonas/genética , Humanos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Ligando Inductor de Apoptosis Relacionado con TNF/genética
17.
Cell Stem Cell ; 19(1): 66-80, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-27320042

RESUMEN

The RNA-binding proteins LIN28A and LIN28B play critical roles in embryonic development, tumorigenesis, and pluripotency, but their exact functions are poorly understood. Here, we show that, like LIN28A, LIN28B can function effectively with NANOG, OCT4, and SOX2 in reprogramming to pluripotency and that reactivation of both endogenous LIN28A and LIN28B loci are required for maximal reprogramming efficiency. In human fibroblasts, LIN28B is activated early during reprogramming, while LIN28A is activated later during the transition to bona fide induced pluripotent stem cells (iPSCs). In murine cells, LIN28A and LIN28B facilitate conversion from naive to primed pluripotency. Proteomic and metabolomic analysis highlighted roles for LIN28 in maintaining the low mitochondrial function associated with primed pluripotency and in regulating one-carbon metabolism, nucleotide metabolism, and histone methylation. LIN28 binds to mRNAs of proteins important for oxidative phosphorylation and modulates protein abundance. Thus, LIN28A and LIN28B play cooperative roles in regulating reprogramming, naive/primed pluripotency, and stem cell metabolism.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Carbono/metabolismo , Reprogramación Celular , Fibroblastos/metabolismo , Histonas/metabolismo , Humanos , Metilación , Ratones , Nucleótidos/metabolismo , Oxidación-Reducción , Fosforilación Oxidativa , Proteoma/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
18.
Methods Mol Biol ; 1353: 215-31, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26126451

RESUMEN

Induced pluripotent stem cells (iPSCs) offer great promise as tools for basic biomedical research, disease modeling, and drug screening. In this chapter, we describe the generation of patient-specific, transgene-free iPSCs from skin biopsies and peripheral blood mononuclear cells through electroporation of episomal vectors and growth under two different culture conditions. The resulting iPSC lines are characterized with respect to pluripotency marker expression through immunostaining, tested for transgene integration by PCR, and assayed for differentiation capacity via teratoma formation.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Reprogramación Celular , Fibroblastos/citología , Células Madre Pluripotentes Inducidas/citología , Leucocitos Mononucleares/citología , Amidas/farmacología , Animales , Biomarcadores/metabolismo , Biopsia , Diferenciación Celular/efectos de los fármacos , Colágeno/química , Criopreservación , Combinación de Medicamentos , Electroporación , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Laminina/química , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Ratones , Plásmidos/genética , Plásmidos/metabolismo , Cultivo Primario de Células , Proteoglicanos/química , Piridinas/farmacología , Piel/citología , Piel/metabolismo , Teratoma/genética , Teratoma/metabolismo , Teratoma/patología , Transgenes
19.
Mol Cell ; 53(1): 32-48, 2014 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-24361252

RESUMEN

Self-renewal and pluripotency of embryonic stem cells (ESCs) are established by multiple regulatory pathways operating at several levels. The roles of histone demethylases (HDMs) in these programs are incompletely defined. We conducted a functional RNAi screen for HDMs and identified five potential HDMs essential for mouse ESC identity. In-depth analyses demonstrate that the closely related HDMs Jmjd2b and Jmjd2c are necessary for self-renewal of ESCs and induced pluripotent stem cell generation. Genome-wide occupancy studies reveal that Jmjd2b unique, Jmjd2c unique, and Jmjd2b-Jmjd2c common target sites belong to functionally separable Core, Polycomb repressive complex (PRC), and Myc regulatory modules, respectively. Jmjd2b and Nanog act through an interconnected regulatory loop, whereas Jmjd2c assists PRC2 in transcriptional repression. Thus, two HDMs of the same subclass exhibit distinct and combinatorial functions in control of the ESC state. Such complexity of HDM function reveals an aspect of multilayered transcriptional control.


Asunto(s)
Células Madre Embrionarias/enzimología , Histona Demetilasas con Dominio de Jumonji/metabolismo , Células Madre Pluripotentes/enzimología , Transcripción Genética/fisiología , Animales , Línea Celular , Células Madre Embrionarias/citología , Estudio de Asociación del Genoma Completo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Histona Demetilasas con Dominio de Jumonji/genética , Ratones , Proteína Homeótica Nanog , Células Madre Pluripotentes/citología , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo
20.
Science ; 339(6116): 222-6, 2013 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-23118012

RESUMEN

Threonine is the only amino acid critically required for the pluripotency of mouse embryonic stem cells (mESCs), but the detailed mechanism remains unclear. We found that threonine and S-adenosylmethionine (SAM) metabolism are coupled in pluripotent stem cells, resulting in regulation of histone methylation. Isotope labeling of mESCs revealed that threonine provides a substantial fraction of both the cellular glycine and the acetyl-coenzyme A (CoA) needed for SAM synthesis. Depletion of threonine from the culture medium or threonine dehydrogenase (Tdh) from mESCs decreased accumulation of SAM and decreased trimethylation of histone H3 lysine 4 (H3K4me3), leading to slowed growth and increased differentiation. Thus, abundance of SAM appears to influence H3K4me3, providing a possible mechanism by which modulation of a metabolic pathway might influence stem cell fate.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Histonas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes/metabolismo , S-Adenosilmetionina/metabolismo , Treonina/metabolismo , Acetilcoenzima A/metabolismo , Oxidorreductasas de Alcohol/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Reprogramación Celular , Medios de Cultivo , Epigénesis Genética , Fibroblastos/citología , Fibroblastos/metabolismo , Glicina/metabolismo , Células Madre Pluripotentes Inducidas/citología , Redes y Vías Metabólicas , Metilación , Ratones , Células Madre Pluripotentes/citología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA