Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 42(2): e111673, 2023 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-36514940

RESUMEN

Adenosine N6-methylation (m6A) and N6,2'-O-dimethylation (m6Am) are regulatory modifications of eukaryotic mRNAs. m6Am formation is catalyzed by the methyl transferase phosphorylated CTD-interacting factor 1 (PCIF1); however, the pathophysiological functions of this RNA modification and PCIF1 in cancers are unclear. Here, we show that PCIF1 expression is upregulated in colorectal cancer (CRC) and negatively correlates with patient survival. CRISPR/Cas9-mediated depletion of PCIF1 in human CRC cells leads to loss of cell migration, invasion, and colony formation in vitro and loss of tumor growth in athymic mice. Pcif1 knockout in murine CRC cells inhibits tumor growth in immunocompetent mice and enhances the effects of anti-PD-1 antibody treatment by decreasing intratumoral TGF-ß levels and increasing intratumoral IFN-γ, TNF-α levels, and tumor-infiltrating natural killer cells. We further show that PCIF1 modulates CRC growth and response to anti-PD-1 in a context-dependent mechanism with PCIF1 directly targeting FOS, IFITM3, and STAT1 via m6Am modifications. PCIF1 stabilizes FOS mRNA, which in turn leads to FOS-dependent TGF-ß regulation and tumor growth. While during immunotherapy, Pcif1-Fos-TGF-ß, as well as Pcif1-Stat1/Ifitm3-IFN-γ axes, contributes to the resistance of anti-PD-1 therapy. Collectively, our findings reveal a role of PCIF1 in promoting CRC tumorigenesis and resistance to anti-PD-1 therapy, supporting that the combination of PCIF1 inhibition with anti-PD-1 treatment is a potential therapeutic strategy to enhance CRC response to immunotherapy. Finally, we developed a lipid nanoparticles (LNPs) and chemically modified small interfering RNAs (CMsiRNAs)-based strategy to silence PCIF1 in vivo and found that this treatment significantly reduced tumor growth in mice. Our results therefore provide a proof-of-concept for tumor growth suppression using LNP-CMsiRNA to silence target genes in cancer.


Asunto(s)
Neoplasias Colorrectales , Inmunoterapia , Animales , Humanos , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Proteínas de la Membrana/metabolismo , Metilación , Proteínas Nucleares/metabolismo , ARN Mensajero/genética , Proteínas de Unión al ARN/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
2.
Proc Natl Acad Sci U S A ; 120(5): e2210361120, 2023 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-36689652

RESUMEN

Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to be a major health problem worldwide. Due to the fast emergence of SARS-CoV-2 variants, understanding the molecular mechanisms of viral pathogenesis and developing novel inhibitors are essential and urgent. Here, we investigated the potential roles of N6,2'-O-dimethyladenosine (m6Am), one of the most abundant modifications of eukaryotic messenger ribonucleic acid (mRNAs), in SARS-CoV-2 infection of human cells. Using genome-wide m6Am-exo-seq, RNA sequencing analysis, and Clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 genome editing, we demonstrate that phosphorylated C-terminal domain (CTD)-interacting factor 1 (PCIF1), a cap-specific adenine N6-methyltransferase, plays a major role in facilitating infection of primary human lung epithelial cells and cell lines by SARS-CoV-2, variants of concern, and other coronaviruses. We show that PCIF1 promotes infection by sustaining expression of the coronavirus receptors angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2) via m6Am-dependent mRNA stabilization. In PCIF1-depleted cells, both ACE2/TMPRSS2 expression and viral infection are rescued by re-expression of wild-type, but not catalytically inactive, PCIF1. These findings suggest a role for PCIF1 and cap m6Am in regulating SARS-CoV-2 susceptibility and identify a potential therapeutic target for prevention of infection.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2/genética , Enzima Convertidora de Angiotensina 2 , ARN Mensajero/genética , Proteínas Nucleares/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Serina Endopeptidasas
3.
EMBO Rep ; 24(10): e55506, 2023 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-37705505

RESUMEN

N6 -methyladenosine (m6 A), the most abundant internal modification in eukaryotic mRNA, plays important roles in many physiological and pathological processes, including the development and progression of cancer. RNA modification by m6 A is regulated by methyltransferases, demethylases, and m6 A-binding proteins that function in large part by regulating mRNA expression and function. Here, we investigate the expression of m6 A regulatory proteins in breast cancer. We find that expression of KIAA1429/VIRMA, a component of the m6 A methyltransferase complex, is upregulated in breast cancer tissue and correlates positively with poor survival. KIAA1429/VIRMA is mislocalized to the cytosol of breast cancer tissues and cell lines, and shRNA-mediated knockdown inhibits breast cancer cell proliferation, migration, and invasion. Mechanistically, KIAA1429/VIRMA is shown to bind to the m6 A-dependent RNA-binding protein insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3), leading to recruitment and stabilization of m6 A-modified hyaluronan synthase 2 (HAS2) mRNA. HAS2 mRNA and KIAA1429/VIRMA mRNA levels correlate positively in breast cancer tissues, suggesting that the KIAA1429/VIRMA-IGF2BP3-HAS2 axis promotes breast cancer growth and contributes to poor prognosis.


Asunto(s)
Neoplasias , Humanos , Citosol , Hialuronano Sintasas , Citoplasma , ARN Mensajero/genética
4.
EMBO J ; 39(20): e104514, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32964498

RESUMEN

An impressive clinical success has been observed in treating a variety of cancers using immunotherapy with programmed cell death-1 (PD-1) checkpoint blockade. However, limited response in most patients treated with anti-PD-1 antibodies remains a challenge, requiring better understanding of molecular mechanisms limiting immunotherapy. In colorectal cancer (CRC) resistant to immunotherapy, mismatch-repair-proficient or microsatellite instability-low (pMMR-MSI-L) tumors have low mutation burden and constitute ~85% of patients. Here, we show that inhibition of N6 -methyladenosine (m6 A) mRNA modification by depletion of methyltransferases, Mettl3 and Mettl14, enhanced response to anti-PD-1 treatment in pMMR-MSI-L CRC and melanoma. Mettl3- or Mettl14-deficient tumors increased cytotoxic tumor-infiltrating CD8+ T cells and elevated secretion of IFN-γ, Cxcl9, and Cxcl10 in tumor microenvironment in vivo. Mechanistically, Mettl3 or Mettl14 loss promoted IFN-γ-Stat1-Irf1 signaling through stabilizing the Stat1 and Irf1 mRNA via Ythdf2. Finally, we found a negative correlation between METTL3 or METTL14 and STAT1 in 59 patients with pMMR-MSI-L CRC tumors. Altogether, our findings uncover a new awareness of the function of RNA methylation in adaptive immunity and provide METTL3 and METTL14 as potential therapeutic targets in anticancer immunotherapy.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/enzimología , Citocinas/metabolismo , Inmunoterapia/métodos , Melanoma/enzimología , Metiltransferasas/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Proliferación Celular/genética , Quimiocina CXCL10 , Quimiocina CXCL9/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , Humanos , Inhibidores de Puntos de Control Inmunológico , Inmunohistoquímica , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/metabolismo , Interferón gamma/metabolismo , Melanoma/genética , Melanoma/inmunología , Melanoma/metabolismo , Metiltransferasas/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , RNA-Seq , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Microambiente Tumoral/genética
5.
EMBO J ; 39(21): e106057, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32944968

RESUMEN

Coronavirus disease 2019 (COVID-19) is caused by SARS-CoV-2 and has spread across the globe. SARS-CoV-2 is a highly infectious virus with no vaccine or antiviral therapy available to control the pandemic; therefore, it is crucial to understand the mechanisms of viral pathogenesis and the host immune responses to SARS-CoV-2. SARS-CoV-2 is a new member of the betacoronavirus genus like other closely related viruses including SARS-CoV and Middle East respiratory syndrome coronavirus (MERS-CoV). Both SARS-CoV and MERS-CoV have caused serious outbreaks and epidemics in the past eighteen years. Here, we report that one of the interferon-stimulated genes (ISGs), cholesterol 25-hydroxylase (CH25H), is induced by SARS-CoV-2 infection in vitro and in COVID-19-infected patients. CH25H converts cholesterol to 25-hydrocholesterol (25HC) and 25HC shows broad anti-coronavirus activity by blocking membrane fusion. Furthermore, 25HC inhibits USA-WA1/2020 SARS-CoV-2 infection in lung epithelial cells and viral entry in human lung organoids. Mechanistically, 25HC inhibits viral membrane fusion by activating the ER-localized acyl-CoA:cholesterol acyltransferase (ACAT) which leads to the depletion of accessible cholesterol from the plasma membrane. Altogether, our results shed light on a potentially broad antiviral mechanism by 25HC through depleting accessible cholesterol on the plasma membrane to suppress virus-cell fusion. Since 25HC is a natural product with no known toxicity at effective concentrations, it provides a potential therapeutic candidate for COVID-19 and emerging viral diseases in the future.


Asunto(s)
Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Colesterol/metabolismo , Infecciones por Coronavirus/tratamiento farmacológico , Neumonía Viral/tratamiento farmacológico , Mucosa Respiratoria/virología , Esteroide Hidroxilasas/farmacología , Internalización del Virus/efectos de los fármacos , Acetil-CoA C-Acetiltransferasa/metabolismo , Animales , COVID-19 , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Chlorocebus aethiops , Activación Enzimática/efectos de los fármacos , Humanos , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Organoides/virología , Pandemias , Mucosa Respiratoria/efectos de los fármacos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , SARS-CoV-2 , Células Vero , Tratamiento Farmacológico de COVID-19
6.
Nucleic Acids Res ; 50(12): 6687-6701, 2022 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-35713529

RESUMEN

The retrovirus human immunodeficiency virus-1 (HIV-1) is the causative agent of AIDS. Although treatment of HIV/AIDS with antiretroviral therapy provides suppression of viremia, latent reservoirs of integrated proviruses preclude cure by current antiviral treatments. Understanding the mechanisms of host-viral interactions may elucidate new treatment strategies. Here, we performed a CRISPR/Cas9 transcriptional activation screen using a high-complexity, genome-wide sgRNA library to identify cellular factors that inhibit HIV-1 infection of human CD4+ T cells. MT4 cells were transduced with a CRISPR/Cas9 sgRNA library and infected with nef-deficient HIV-1NL4-3 expressing ganciclovir-sensitive thymidine kinase, thus enabling selection of HIV-1-resistant cells for analysis of enriched sgRNAs. After validation of screen hits, multiple host factors essential for HIV-1 infection were identified, including SET (SET nuclear proto-oncogene) and ANP32A (acidic nuclear phosphoprotein 32A, PP32A), which together form a histone acetylase inhibitor complex. Using multiple human cell lines and peripheral blood mononuclear cells (PBMCs) from healthy donors and HIV-1-infected individuals, we demonstrate that SET depletion increased HIV-1 infectivity by augmenting DNA integration without significantly changing sites of integration. Conversely, SET overexpression decreased HIV-1 integration and infectivity. SET protein expression was significantly reduced in PBMCs from HIV-1-infected individuals and was downregulated by HIV-1 infection of healthy donor cells in vitro. Notably, HIV-1-induced downregulation of SET could be alleviated by inhibition of the protease granzyme A. Altogether, we have identified cellular inhibitors of HIV-1 infection on a genome-wide scale, which affords new insight into host-virus interactions and may provide new strategies for HIV-1 treatment.


Asunto(s)
VIH-1 , Humanos , Sistemas CRISPR-Cas , Histona Acetiltransferasas , VIH-1/genética , Leucocitos Mononucleares , Proteínas Nucleares , Proteínas de Unión al ARN , Activación Transcripcional , Integración Viral
7.
Genes Dev ; 30(1): 117-31, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26728557

RESUMEN

The transcription cycle of RNA polymerase II (Pol II) is regulated at discrete transition points by cyclin-dependent kinases (CDKs). Positive transcription elongation factor b (P-TEFb), a complex of Cdk9 and cyclin T1, promotes release of paused Pol II into elongation, but the precise mechanisms and targets of Cdk9 action remain largely unknown. Here, by a chemical genetic strategy, we identified ∼ 100 putative substrates of human P-TEFb, which were enriched for proteins implicated in transcription and RNA catabolism. Among the RNA processing factors phosphorylated by Cdk9 was the 5'-to-3' "torpedo" exoribonuclease Xrn2, required in transcription termination by Pol II, which we validated as a bona fide P-TEFb substrate in vivo and in vitro. Phosphorylation by Cdk9 or phosphomimetic substitution of its target residue, Thr439, enhanced enzymatic activity of Xrn2 on synthetic substrates in vitro. Conversely, inhibition or depletion of Cdk9 or mutation of Xrn2-Thr439 to a nonphosphorylatable Ala residue caused phenotypes consistent with inefficient termination in human cells: impaired Xrn2 chromatin localization and increased readthrough transcription of endogenous genes. Therefore, in addition to its role in elongation, P-TEFb regulates termination by promoting chromatin recruitment and activation of a cotranscriptional RNA processing enzyme, Xrn2.


Asunto(s)
Quinasa 9 Dependiente de la Ciclina/metabolismo , Exorribonucleasas/genética , Exorribonucleasas/metabolismo , Regulación de la Expresión Génica/genética , Factor B de Elongación Transcripcional Positiva/metabolismo , Cromatina/metabolismo , Activación Enzimática/genética , Pruebas Genéticas , Células HCT116 , Humanos , Fosforilación , Unión Proteica
8.
Clin Infect Dis ; 76(3): e530-e532, 2023 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35723411

RESUMEN

We isolated a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) BA.2 variant from a person with coronavirus disease 2019 recrudescence after nirmatrelvir/ritonavir treatment. Antiviral sensitivity and neutralizing antibody testing were performed with both parental SARS-CoV-2 and multiple variants of concern. We found that neither nirmatrelvir resistance nor absence of neutralizing immunity was a likely cause of the recrudescence.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2 , Ritonavir/uso terapéutico , Tratamiento Farmacológico de COVID-19
9.
EMBO J ; 38(8)2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30918008

RESUMEN

Long noncoding RNAs (lncRNAs) can regulate target gene expression by acting in cis (locally) or in trans (non-locally). Here, we performed genome-wide expression analysis of Toll-like receptor (TLR)-stimulated human macrophages to identify pairs of cis-acting lncRNAs and protein-coding genes involved in innate immunity. A total of 229 gene pairs were identified, many of which were commonly regulated by signaling through multiple TLRs and were involved in the cytokine responses to infection by group B Streptococcus We focused on elucidating the function of one lncRNA, named lnc-MARCKS or ROCKI (Regulator of Cytokines and Inflammation), which was induced by multiple TLR stimuli and acted as a master regulator of inflammatory responses. ROCKI interacted with APEX1 (apurinic/apyrimidinic endodeoxyribonuclease 1) to form a ribonucleoprotein complex at the MARCKS promoter. In turn, ROCKI-APEX1 recruited the histone deacetylase HDAC1, which removed the H3K27ac modification from the promoter, thus reducing MARCKS transcription and subsequent Ca2+ signaling and inflammatory gene expression. Finally, genetic variants affecting ROCKI expression were linked to a reduced risk of certain inflammatory and infectious disease in humans, including inflammatory bowel disease and tuberculosis. Collectively, these data highlight the importance of cis-acting lncRNAs in TLR signaling, innate immunity, and pathophysiological inflammation.


Asunto(s)
Regulación de la Expresión Génica , Inmunidad Innata/inmunología , Inflamación/inmunología , Macrófagos/inmunología , ARN Largo no Codificante/metabolismo , Infecciones Estreptocócicas/microbiología , Receptores Toll-Like/metabolismo , Células Cultivadas , Citocinas/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Genoma Humano , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Humanos , Inflamación/genética , Inflamación/microbiología , Macrófagos/metabolismo , Macrófagos/microbiología , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada/genética , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada/metabolismo , Regiones Promotoras Genéticas , ARN Largo no Codificante/genética , Infecciones Estreptocócicas/inmunología , Streptococcus agalactiae/aislamiento & purificación , Receptores Toll-Like/genética
10.
Proc Natl Acad Sci U S A ; 117(33): 20159-20170, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32747553

RESUMEN

Although immune checkpoint blockade (ICB) therapy has revolutionized cancer treatment, many patients do not respond or develop resistance to ICB. N6 -methylation of adenosine (m6A) in RNA regulates many pathophysiological processes. Here, we show that deletion of the m6A demethylase Alkbh5 sensitized tumors to cancer immunotherapy. Alkbh5 has effects on m6A density and splicing events in tumors during ICB. Alkbh5 modulates Mct4/Slc16a3 expression and lactate content of the tumor microenvironment and the composition of tumor-infiltrating Treg and myeloid-derived suppressor cells. Importantly, a small-molecule Alkbh5 inhibitor enhanced the efficacy of cancer immunotherapy. Notably, the ALKBH5 gene mutation and expression status of melanoma patients correlate with their response to immunotherapy. Our results suggest that m6A demethylases in tumor cells contribute to the efficacy of immunotherapy and identify ALKBH5 as a potential therapeutic target to enhance immunotherapy outcome in melanoma, colorectal, and potentially other cancers.


Asunto(s)
Desmetilasa de ARN, Homólogo 5 de AlkB/metabolismo , Vacunas contra el Cáncer/inmunología , Lactatos/metabolismo , Melanoma/metabolismo , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T Reguladores/fisiología , Desmetilasa de ARN, Homólogo 5 de AlkB/genética , Anticuerpos , Citocinas/genética , Citocinas/metabolismo , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/terapia , Metiltransferasas/genética , Metiltransferasas/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Células Supresoras de Origen Mieloide/fisiología , Sitios de Empalme de ARN , Empalme del ARN , Simportadores/genética , Simportadores/metabolismo , Transcriptoma , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
Mol Psychiatry ; 26(4): 1194-1207, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32051547

RESUMEN

Methamphetamine (METH) is a potent stimulant that induces a euphoric state but also causes cognitive impairment, neurotoxicity and neurodevelopmental deficits. Yet, the molecular mechanisms by which METH causes neurodevelopmental defects have remained elusive. Here we utilized human cerebral organoids and single-cell RNA sequencing (scRNA-seq) to study the effects of prenatal METH exposure on fetal brain development. We analyzed 20,758 cells from eight untreated and six METH-treated cerebral organoids and found that the organoids developed from embryonic stem cells contained a diverse array of glial and neuronal cell types. We further identified transcriptionally distinct populations of astrocytes and oligodendrocytes within cerebral organoids. Treatment of organoids with METH-induced marked changes in transcription in multiple cell types, including astrocytes and neural progenitor cells. METH also elicited novel astrocyte-specific gene expression networks regulating responses to cytokines, and inflammasome. Moreover, upregulation of immediate early genes, complement factors, apoptosis, and immune response genes suggests a neuroinflammatory program induced by METH regulating neural stem cell proliferation, differentiation, and cell death. Finally, we observed marked METH-induced changes in neuroinflammatory and cytokine gene expression at the RNA and protein levels. Our data suggest that human cerebral organoids represent a model system to study drug-induced neuroinflammation at single-cell resolution.


Asunto(s)
Estimulantes del Sistema Nervioso Central , Metanfetamina , Astrocitos , Humanos , Neuronas , Organoides
12.
EMBO Rep ; 21(12): e49183, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33073500

RESUMEN

Zika virus (ZIKV) is an emerging flavivirus, which when passed through vertical transmission from mother to developing fetus can lead to developmental abnormalities, including microcephaly. While there is mounting evidence that suggests a causal relationship between ZIKV infection and microcephaly, the mechanisms by which ZIKV induces these changes remain to be elucidated. Here, we demonstrate that ZIKV infection of neural stems cells, both in vitro and in vivo, induces macroautophagy to enhance viral replication. At the same time, ZIKV downregulates a number of essential selective autophagy genes, including the Fanconi anemia (FA) pathway genes. Bioinformatics analyses indicate that the transcription factor E2F4 promotes FANCC expression and is downregulated upon ZIKV infection. Gain and loss of function assays indicate that FANCC is essential for selective autophagy and acts as a negative regulator of ZIKV replication. Finally, we show that Fancc KO mice have increased ZIKV infection and autophagy protein levels in various brain regions. Taken together, ZIKV downregulates FANCC to modulate the host antiviral response and simultaneously attenuate neuronal growth.


Asunto(s)
Anemia de Fanconi , Células-Madre Neurales , Infección por el Virus Zika , Virus Zika , Animales , Autofagia , Línea Celular , Anemia de Fanconi/genética , Proteína del Grupo de Complementación C de la Anemia de Fanconi , Macroautofagia , Ratones , Replicación Viral , Virus Zika/genética , Infección por el Virus Zika/genética
13.
Mol Cell ; 53(6): 1005-19, 2014 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-24530304

RESUMEN

Here, we generated a genome-scale shRNA library targeting long intergenic noncoding RNAs (lincRNAs) in the mouse. We performed an unbiased loss-of-function study in mouse embryonic stem cells (mESCs) and identified 20 lincRNAs involved in the maintenance of pluripotency. Among these, TUNA (Tcl1 Upstream Neuron-Associated lincRNA, or megamind) was required for pluripotency and formed a complex with three RNA-binding proteins (RBPs). The TUNA-RBP complex was detected at the promoters of Nanog, Sox2, and Fgf4, and knockdown of TUNA or the individual RBPs inhibited neural differentiation of mESCs. TUNA showed striking evolutionary conservation of both sequence- and CNS-restricted expression in vertebrates. Accordingly, knockdown of tuna in zebrafish caused impaired locomotor function, and TUNA expression in the brains of Huntington's disease patients was significantly associated with disease grade. Our results suggest that the lincRNA TUNA plays a vital role in pluripotency and neural differentiation of ESCs and is associated with neurological function of adult vertebrates.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Enfermedad de Huntington/genética , Neuronas/metabolismo , Células Madre Pluripotentes/metabolismo , ARN Largo no Codificante/genética , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Evolución Biológica , Diferenciación Celular , Secuencia Conservada , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Factor 4 de Crecimiento de Fibroblastos/genética , Factor 4 de Crecimiento de Fibroblastos/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Ratones , Datos de Secuencia Molecular , Actividad Motora , Proteína Homeótica Nanog , Neuronas/citología , Células Madre Pluripotentes/citología , Regiones Promotoras Genéticas , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Homología de Secuencia de Aminoácido , Índice de Severidad de la Enfermedad , Transducción de Señal , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
14.
Small ; 17(15): e2006050, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33502104

RESUMEN

Glioblastoma multiforme (GBM) is the most lethal primary brain tumor characterized by high cellular and molecular heterogeneity, hypervascularization, and innate drug resistance. Cellular components and extracellular matrix (ECM) are the two primary sources of heterogeneity in GBM. Here, biomimetic tri-regional GBM models with tumor regions, acellular ECM regions, and an endothelial region with regional stiffnesses patterned corresponding to the GBM stroma, pathological or normal brain parenchyma, and brain capillaries, are developed. Patient-derived GBM cells, human endothelial cells, and hyaluronic acid derivatives are used to generate a species-matched and biochemically relevant microenvironment. This in vitro study demonstrates that biophysical cues are involved in various tumor cell behaviors and angiogenic potentials and promote different molecular subtypes of GBM. The stiff models are enriched in the mesenchymal subtype, exhibit diffuse invasion of tumor cells, and induce protruding angiogenesis and higher drug resistance to temozolomide. Meanwhile, the soft models demonstrate enrichment in the classical subtype and support expansive cell growth. The three-dimensional bioprinting technology utilized in this study enables rapid, flexible, and reproducible patient-specific GBM modeling with biophysical heterogeneity that can be employed by future studies as a tunable system to interrogate GBM disease mechanisms and screen drug compounds.


Asunto(s)
Bioimpresión , Neoplasias Encefálicas , Glioblastoma , Línea Celular Tumoral , Células Endoteliales , Humanos , Microambiente Tumoral
15.
PLoS Pathog ; 12(11): e1006034, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27893816

RESUMEN

microRNAs are endogenous small regulatory RNAs that modulate myriad biological processes by repressing target gene expression in a sequence-specific manner. Here we show that the conserved miRNA miR-34 regulates innate immunity and ecdysone signaling in Drosophila. miR-34 over-expression activates antibacterial innate immunity signaling both in cultured cells and in vivo, and flies over-expressing miR-34 display improved survival and pathogen clearance upon Gram-negative bacterial infection; whereas miR-34 knockout animals are defective in antibacterial defense. In particular, miR-34 achieves its immune-stimulatory function, at least in part, by repressing the two novel target genes Dlg1 and Eip75B. In addition, our study reveals a mutual repression between miR-34 expression and ecdysone signaling, and identifies miR-34 as a node in the intricate interplay between ecdysone signaling and innate immunity. Lastly, we identify cis-regulatory genomic elements and trans-acting transcription factors required for optimal ecdysone-mediated repression of miR-34. Taken together, our study enriches the repertoire of immune-modulating miRNAs in animals, and provides new insights into the interplay between steroid hormone signaling and innate immunity.


Asunto(s)
Drosophila melanogaster/inmunología , Ecdisona/inmunología , Inmunidad Innata/inmunología , MicroARNs/inmunología , Transducción de Señal , Animales , Northern Blotting , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/inmunología , Modelos Animales de Enfermedad , Proteínas de Drosophila/inmunología , Técnicas de Inactivación de Genes , Inmunoprecipitación , Reacción en Cadena de la Polimerasa , Transducción de Señal/inmunología , Factores de Transcripción/inmunología , Proteínas Supresoras de Tumor/inmunología
16.
EMBO Rep ; 17(12): 1814-1828, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27797851

RESUMEN

Hematopoietic stem cells (HSCs) are capable of giving rise to all blood cell lineages throughout adulthood, and the generation of engraftable HSCs from human pluripotent stem cells is a major goal for regenerative medicine. Here, we describe a functional genome-wide RNAi screen to identify genes required for the differentiation of embryonic stem cell (ESC) into hematopoietic stem/progenitor cells (HSPCs) in vitro We report the discovery of novel genes important for the endothelial-to-hematopoietic transition and subsequently for HSPC specification. High-throughput sequencing and bioinformatic analyses identified twelve groups of genes, including a set of 351 novel genes required for HSPC specification. As in vivo proof of concept, four of these genes, Ap2a1, Mettl22, Lrsam1, and Hal, are selected for validation, confirmed to be essential for HSPC development in zebrafish and for maintenance of human HSCs. Taken together, our results not only identify a number of novel regulatory genes and pathways essential for HSPC development but also serve as valuable resource for directed differentiation of therapy grade HSPCs using human pluripotent stem cells.


Asunto(s)
Células Madre Embrionarias/fisiología , Hematopoyesis , Células Madre Hematopoyéticas/fisiología , Redes y Vías Metabólicas/genética , Animales , Linaje de la Célula/genética , Linaje de la Célula/fisiología , Células Cultivadas , Biología Computacional , Sangre Fetal/citología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Interferencia de ARN , Transducción de Señal/genética , Pez Cebra/genética
17.
Mol Cell ; 34(6): 696-709, 2009 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-19560422

RESUMEN

MicroRNAs (miRNAs), approximately 22 nt noncoding RNAs, assemble into RNA-induced silencing complexes (RISCs) and localize to cytoplasmic substructures called P bodies. Dictated by base-pair complementarity between miRNA and a target mRNA, miRNAs specifically repress posttranscriptional expression of several mRNAs. Here we report that HIV-1 mRNA interacts with RISC proteins and that disrupting P body structures enhances viral production and infectivity. In HIV-1-infected human T lymphocytes, we identified a highly abundant miRNA, miR-29a, which specifically targets the HIV-1 3'UTR region. Inhibiting miR-29a enhanced HIV-1 viral production and infectivity, whereas expressing a miR-29 mimic suppressed viral replication. We also found that specific miR-29a-HIV-1 mRNA interactions enhance viral mRNA association with RISC and P body proteins. Thus we provide an example of a single host miRNA regulating HIV-1 production and infectivity. These studies highlight the significance of miRNAs and P bodies in modulating host cell interactions with HIV-1 and possibly other viruses.


Asunto(s)
VIH-1/patogenicidad , MicroARNs/fisiología , ARN Mensajero/fisiología , ARN Viral/fisiología , Complejo Silenciador Inducido por ARN/metabolismo , Secuencia de Bases , Sitios de Unión , Estructuras Citoplasmáticas/fisiología , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , VIH-1/genética , VIH-1/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Datos de Secuencia Molecular , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/química , ARN Viral/química , Ribonucleasa III/antagonistas & inhibidores , Linfocitos T/virología , Replicación Viral
18.
Proc Natl Acad Sci U S A ; 111(3): 1002-7, 2014 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-24371310

RESUMEN

Thousands of large intergenic noncoding RNAs (lincRNAs) have been identified in the mammalian genome, many of which have important roles in regulating a variety of biological processes. Here, we used a custom microarray to identify lincRNAs associated with activation of the innate immune response. A panel of 159 lincRNAs was found to be differentially expressed following innate activation of THP1 macrophages. Among them, linc1992 was shown to be expressed in many human tissues and was required for induction of TNFα expression. Linc1992 bound specifically to heterogenous nuclear ribonucleoprotein L (hnRNPL) and formed a functional linc1992-hnRNPL complex that regulated transcription of the TNFα gene by binding to its promoter. Transcriptome analysis revealed that linc1992 was required for expression of many immune-response genes, including other cytokines and transcriptional and posttranscriptional regulators of TNFα expression, and that knockdown of linc1992 caused dysregulation of these genes during innate activation of THP1 macrophages. Therefore, we named linc1992 THRIL (TNFα and hnRNPL related immunoregulatory LincRNA). Finally, THRIL expression was correlated with the severity of symptoms in patients with Kawasaki disease, an acute inflammatory disease of childhood. Collectively, our data provide evidence that lincRNAs and their binding proteins can regulate TNFα expression and may play important roles in the innate immune response and inflammatory diseases in humans.


Asunto(s)
Regulación de la Expresión Génica , Ribonucleoproteína Heterogénea-Nuclear Grupo L/metabolismo , ARN Largo no Codificante/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Línea Celular , Nucléolo Celular/metabolismo , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Perfilación de la Expresión Génica , Humanos , Inmunidad Innata , Inflamación , Interleucina-6/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Síndrome Mucocutáneo Linfonodular/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos
19.
Immunol Rev ; 253(1): 97-111, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23550641

RESUMEN

RNA interference (RNAi) is an ancient process by which non-coding RNAs regulate gene expression in a sequence-specific manner. The core components of RNAi are small regulatory RNAs, approximately 21-30 nucleotides in length, including small interfering RNAs (siRNAs) and microRNAs (miRNAs). The past two decades have seen considerable progress in our understanding of the molecular mechanisms underlying the biogenesis of siRNAs and miRNAs. Recent advances have also revealed the crucial regulatory roles played by small RNAs in such diverse processes as development, homeostasis, innate immunity, and oncogenesis. Accumulating evidence indicates that RNAi initially evolved as a host defense mechanism against viruses and transposons. The ability of the host small RNA biogenesis machinery to recognize viral double-stranded RNA replication intermediates and transposon transcripts is critical to this process, as is small RNA-guided targeting of RNAs via complementary base pairing. Collectively, these properties confer unparalleled specificity and precision to RNAi-mediated gene silencing as an effective antiviral mechanism.


Asunto(s)
Interacciones Huésped-Patógeno , ARN no Traducido/inmunología , ARN Viral/inmunología , Virosis/inmunología , Virus/inmunología , Animales , Evolución Biológica , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Interferencia de ARN , Virosis/genética , Virus/genética
20.
Crit Rev Biochem Mol Biol ; 49(1): 16-32, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24164576

RESUMEN

The past two decades have seen an explosion in research on non-coding RNAs and their physiological and pathological functions. Several classes of small (20-30 nucleotides) and long (>200 nucleotides) non-coding RNAs have been firmly established as key regulators of gene expression in myriad processes ranging from embryonic development to innate immunity. In this review, we focus on our current understanding of the molecular mechanisms underlying the biogenesis and function of small interfering RNAs (siRNAs), microRNAs (miRNAs) and Piwi-interacting RNAs (piRNAs). In addition, we briefly review the relevance of small and long non-coding RNAs to human physiology and pathology and their potential to be exploited as therapeutic agents.


Asunto(s)
Regulación de la Expresión Génica , ARN no Traducido/metabolismo , Animales , Impresión Genómica , Humanos , Neoplasias/genética , Interferencia de ARN , ARN no Traducido/genética , ARN no Traducido/uso terapéutico , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA