Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 180
Filtrar
1.
Nat Rev Mol Cell Biol ; 13(4): 239-50, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-22436747

RESUMEN

MicroRNAs (miRNAs) have recently emerged as key regulators of metabolism. For example, miR-33a and miR-33b have a crucial role in controlling cholesterol and lipid metabolism in concert with their host genes, the sterol-regulatory element-binding protein (SREBP) transcription factors. Other metabolic miRNAs, such as miR-103 and miR-107, regulate insulin and glucose homeostasis, whereas miRNAs such as miR-34a are emerging as key regulators of hepatic lipid homeostasis. The discovery of circulating miRNAs has highlighted their potential as both endocrine signalling molecules and disease markers. Dysregulation of miRNAs may contribute to metabolic abnormalities, suggesting that miRNAs may potentially serve as therapeutic targets for ameliorating cardiometabolic disorders.


Asunto(s)
Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , MicroARNs/metabolismo , Colesterol/metabolismo , Sistema Endocrino/metabolismo , Hígado Graso/genética , Hígado Graso/metabolismo , Glucosa/metabolismo , Homeostasis , Humanos , Insulina/metabolismo , Metabolismo de los Lípidos , Síndrome Metabólico/genética , Síndrome Metabólico/metabolismo , MicroARNs/genética , Enfermedad del Hígado Graso no Alcohólico , Obesidad/genética , Obesidad/metabolismo , Oligorribonucleótidos Antisentido/farmacología , Transducción de Señal , Proteínas de Unión a los Elementos Reguladores de Esteroles/genética , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo
2.
Proc Natl Acad Sci U S A ; 113(43): 11998-12005, 2016 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-27790981

RESUMEN

Many Leishmania (Viannia) parasites harbor the double-stranded RNA virus Leishmania RNA virus 1 (LRV1), which has been associated with increased disease severity in animal models and humans and with drug treatment failures in humans. Remarkably, LRV1 survives in the presence of an active RNAi pathway, which in many organisms controls RNA viruses. We found significant levels (0.4 to 2.5%) of small RNAs derived from LRV1 in both Leishmania braziliensis and Leishmania guyanensis, mapping across both strands and with properties consistent with Dicer-mediated cleavage of the dsRNA genome. LRV1 lacks cis- or trans-acting RNAi inhibitory activities, suggesting that virus retention must be maintained by a balance between RNAi activity and LRV1 replication. To tilt this balance toward elimination, we targeted LRV1 using long-hairpin/stem-loop constructs similar to those effective against chromosomal genes. LRV1 was completely eliminated, at high efficiency, accompanied by a massive overproduction of LRV1-specific siRNAs, representing as much as 87% of the total. For both L. braziliensis and L. guyanensis, RNAi-derived LRV1-negative lines were no longer able to induce a Toll-like receptor 3-dependent hyperinflammatory cytokine response in infected macrophages. We demonstrate in vitro a role for LRV1 in virulence of L. braziliensis, the Leishmania species responsible for the vast majority of mucocutaneous leishmaniasis cases. These findings establish a targeted method for elimination of LRV1, and potentially of other Leishmania viruses, which will facilitate mechanistic dissection of the role of LRV1-mediated virulence. Moreover, our data establish a third paradigm for RNAi-viral relationships in evolution: one of balance rather than elimination.


Asunto(s)
Antiprotozoarios/farmacología , Leishmaniasis Mucocutánea/tratamiento farmacológico , Leishmaniavirus/efectos de los fármacos , Oligorribonucleótidos Antisentido/farmacología , ARN Bicatenario/antagonistas & inhibidores , ARN Viral/antagonistas & inhibidores , Animales , Antiprotozoarios/química , Antiprotozoarios/metabolismo , Expresión Génica , Secuencias Invertidas Repetidas , Leishmania braziliensis/patogenicidad , Leishmania braziliensis/virología , Leishmania guyanensis/patogenicidad , Leishmania guyanensis/virología , Leishmaniasis Mucocutánea/parasitología , Leishmaniasis Mucocutánea/virología , Leishmaniavirus/genética , Leishmaniavirus/metabolismo , Macrófagos/parasitología , Macrófagos/virología , Ratones , Oligorribonucleótidos Antisentido/genética , Oligorribonucleótidos Antisentido/metabolismo , Interferencia de ARN/efectos de los fármacos , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Simbiosis/genética , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo , Replicación Viral/efectos de los fármacos
3.
Am J Hum Genet ; 97(4): 555-66, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26411495

RESUMEN

The nuclear pore complex (NPC) is a huge protein complex embedded in the nuclear envelope. It has central functions in nucleocytoplasmic transport, nuclear framework, and gene regulation. Nucleoporin 107 kDa (NUP107) is a component of the NPC central scaffold and is an essential protein in all eukaryotic cells. Here, we report on biallelic NUP107 mutations in nine affected individuals who are from five unrelated families and show early-onset steroid-resistant nephrotic syndrome (SRNS). These individuals have pathologically focal segmental glomerulosclerosis, a condition that leads to end-stage renal disease with high frequency. NUP107 is ubiquitously expressed, including in glomerular podocytes. Three of four NUP107 mutations detected in the affected individuals hamper NUP107 binding to NUP133 (nucleoporin 133 kDa) and NUP107 incorporation into NPCs in vitro. Zebrafish with nup107 knockdown generated by morpholino oligonucleotides displayed hypoplastic glomerulus structures and abnormal podocyte foot processes, thereby mimicking the pathological changes seen in the kidneys of the SRNS individuals with NUP107 mutations. Considering the unique properties of the podocyte (highly differentiated foot-process architecture and slit membrane and the inability to regenerate), we propose a "podocyte-injury model" as the pathomechanism for SRNS due to biallelic NUP107 mutations.


Asunto(s)
Edad de Inicio , Mutación/genética , Síndrome Nefrótico/congénito , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Alelos , Animales , Células Cultivadas , Niño , Preescolar , Citoplasma/metabolismo , Femenino , Haplotipos , Humanos , Immunoblotting , Inmunoprecipitación , Lactante , Riñón/metabolismo , Riñón/patología , Masculino , Microscopía Fluorescente , Síndrome Nefrótico/etiología , Síndrome Nefrótico/patología , Poro Nuclear , Proteínas de Complejo Poro Nuclear/antagonistas & inhibidores , Oligorribonucleótidos Antisentido/farmacología , Linaje , Podocitos/metabolismo , Podocitos/patología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/antagonistas & inhibidores
4.
J Neurosci ; 36(28): 7415-27, 2016 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-27413152

RESUMEN

UNLABELLED: Pathologic inclusions define α-synucleinopathies that include Parkinson's disease (PD). The most common genetic cause of PD is the G2019S LRRK2 mutation that upregulates LRRK2 kinase activity. However, the interaction between α-synuclein, LRRK2, and the formation of α-synuclein inclusions remains unclear. Here, we show that G2019S-LRRK2 expression, in both cultured neurons and dopaminergic neurons in the rat substantia nigra pars compact, increases the recruitment of endogenous α-synuclein into inclusions in response to α-synuclein fibril exposure. This results from the expression of mutant G2019S-LRRK2, as overexpression of WT-LRRK2 not only does not increase formation of inclusions but reduces their abundance. In addition, treatment of primary mouse neurons with LRRK2 kinase inhibitors, PF-06447475 and MLi-2, blocks G2019S-LRRK2 effects, suggesting that the G2019S-LRRK2 potentiation of inclusion formation depends on its kinase activity. Overexpression of G2019S-LRRK2 slightly increases, whereas WT-LRRK2 decreases, total levels of α-synuclein. Knockdown of total α-synuclein with potent antisense oligonucleotides substantially reduces inclusion formation in G2019S-LRRK2-expressing neurons, suggesting that LRRK2 influences α-synuclein inclusion formation by altering α-synuclein levels. These findings support the hypothesis that G2019S-LRRK2 may increase the progression of pathological α-synuclein inclusions after the initial formation of α-synuclein pathology by increasing a pool of α-synuclein that is more susceptible to forming inclusions. SIGNIFICANCE STATEMENT: α-Synuclein inclusions are found in the brains of patients with many different neurodegenerative diseases. Point mutation, duplication, or triplication of the α-synuclein gene can all cause Parkinson's disease (PD). The G2019S mutation in LRRK2 is the most common known genetic cause of PD. The interaction between G2019S-LRRK2 and α-synuclein may uncover new mechanisms and targets for neuroprotection. Here, we show that expression of G2019S-LRRK2 increases α-synuclein mobility and enhances aggregation of α-synuclein in primary cultured neurons and in dopaminergic neurons of the substantia nigra pars compacta, a susceptible brain region in PD. Potent LRRK2 kinase inhibitors, which are being developed for clinical use, block the increased α-synuclein aggregation in G2019S-LRRK2-expressing neurons. These results demonstrate that α-synuclein inclusion formation in neurons can be blocked and that novel therapeutic compounds targeting this process by inhibiting LRRK2 kinase activity may slow progression of PD-associated pathology.


Asunto(s)
Cuerpos de Inclusión/patología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Mutación/genética , Neuronas/metabolismo , Transcitosis/fisiología , alfa-Sinucleína/metabolismo , Animales , Regulación de la Expresión Génica/genética , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oligorribonucleótidos Antisentido/farmacología , Fotoblanqueo , Ratas , Sinucleínas/metabolismo , Transcitosis/genética , Tubulina (Proteína)/metabolismo , Canales Aniónicos Dependientes del Voltaje/genética , Canales Aniónicos Dependientes del Voltaje/metabolismo
5.
Blood ; 126(15): 1844-55, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26286849

RESUMEN

Sickle cell disease (SCD) results in vascular occlusions, chronic hemolytic anemia, and cumulative organ damage. A conspicuous feature of SCD is chronic inflammation and coagulation system activation. Thrombin (factor IIa [FIIa]) is both a central protease in hemostasis and a key modifier of inflammatory processes. To explore the hypothesis that reduced prothrombin (factor II [FII]) levels in SCD will limit vaso-occlusion, vasculopathy, and inflammation, we used 2 strategies to suppress FII in SCD mice. Weekly administration of FII antisense oligonucleotide "gapmer" to Berkeley SCD mice to selectively reduce circulating FII levels to ∼10% of normal for 15 weeks significantly diminished early mortality. More comprehensive, long-term comparative studies were done using mice with genetic diminution of circulating FII. Here, cohorts of FII(lox/-) mice (constitutively carrying ∼10% normal FII) and FII(WT) mice were tracked in parallel for a year following the imposition of SCD via hematopoietic stem cell transplantation. This genetically imposed suppression of FII levels resulted in an impressive reduction in inflammation (reduction in leukocytosis, thrombocytosis, and circulating interleukin-6 levels), reduced endothelial cell dysfunction (reduced endothelial activation and circulating soluble vascular cell adhesion molecule), and a significant improvement in SCD-associated end-organ damage (nephropathy, pulmonary hypertension, pulmonary inflammation, liver function, inflammatory infiltration, and microinfarctions). Notably, all of these benefits were achieved with a relatively modest 1.25-fold increase in prothrombin times, and in the absence of hemorrhagic complications. Taken together, these data establish that prothrombin is a powerful modifier of SCD-induced end-organ damage, and present a novel therapeutic target to ameliorate SCD pathologies.


Asunto(s)
Anemia de Células Falciformes/complicaciones , Terapia Genética , Hipertensión Pulmonar/prevención & control , Inflamación/prevención & control , Protrombina/fisiología , Enfermedades Vasculares/prevención & control , Anemia de Células Falciformes/mortalidad , Anemia de Células Falciformes/fisiopatología , Animales , Coagulación Sanguínea , Células Cultivadas , Hipertensión Pulmonar/etiología , Técnicas para Inmunoenzimas , Inflamación/etiología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Noqueados , Oligorribonucleótidos Antisentido/farmacología , Protrombina/antagonistas & inhibidores , Tasa de Supervivencia , Trombina/metabolismo , Enfermedades Vasculares/etiología
6.
Nature ; 478(7369): 404-7, 2011 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-22012398

RESUMEN

Cardiovascular disease remains the leading cause of mortality in westernized countries, despite optimum medical therapy to reduce the levels of low-density lipoprotein (LDL)-associated cholesterol. The pursuit of novel therapies to target the residual risk has focused on raising the levels of high-density lipoprotein (HDL)-associated cholesterol in order to exploit its atheroprotective effects. MicroRNAs (miRNAs) have emerged as important post-transcriptional regulators of lipid metabolism and are thus a new class of target for therapeutic intervention. MicroRNA-33a and microRNA-33b (miR-33a/b) are intronic miRNAs whose encoding regions are embedded in the sterol-response-element-binding protein genes SREBF2 and SREBF1 (refs 3-5), respectively. These miRNAs repress expression of the cholesterol transporter ABCA1, which is a key regulator of HDL biogenesis. Recent studies in mice suggest that antagonizing miR-33a may be an effective strategy for raising plasma HDL levels and providing protection against atherosclerosis; however, extrapolating these findings to humans is complicated by the fact that mice lack miR-33b, which is present only in the SREBF1 gene of medium and large mammals. Here we show in African green monkeys that systemic delivery of an anti-miRNA oligonucleotide that targets both miR-33a and miR-33b increased hepatic expression of ABCA1 and induced a sustained increase in plasma HDL levels over 12 weeks. Notably, miR-33 antagonism in this non-human primate model also increased the expression of miR-33 target genes involved in fatty acid oxidation (CROT, CPT1A, HADHB and PRKAA1) and reduced the expression of genes involved in fatty acid synthesis (SREBF1, FASN, ACLY and ACACA), resulting in a marked suppression of the plasma levels of very-low-density lipoprotein (VLDL)-associated triglycerides, a finding that has not previously been observed in mice. These data establish, in a model that is highly relevant to humans, that pharmacological inhibition of miR-33a and miR-33b is a promising therapeutic strategy to raise plasma HDL and lower VLDL triglyceride levels for the treatment of dyslipidaemias that increase cardiovascular disease risk.


Asunto(s)
Chlorocebus aethiops , Regulación de la Expresión Génica/efectos de los fármacos , Lipoproteínas HDL/sangre , Lipoproteínas VLDL/sangre , Hígado/efectos de los fármacos , MicroARNs/antagonistas & inhibidores , Oligorribonucleótidos Antisentido/farmacología , Triglicéridos/sangre , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Células Cultivadas , Chlorocebus aethiops/sangre , Chlorocebus aethiops/genética , Chlorocebus aethiops/metabolismo , LDL-Colesterol/sangre , Silenciador del Gen , Células HEK293 , Humanos , Hígado/metabolismo , Masculino , MicroARNs/metabolismo , Factores de Tiempo
7.
Differentiation ; 90(4-5): 91-100, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26677981

RESUMEN

MicroRNAs (miRNAs) are critical in the maintenance, differentiation, and lineage commitment of stem cells. Stem cells have the unique property to differentiate into tissue-specific cell types (lineage commitment) during cell division (self-renewal). In this study, we investigated whether miR-34a, a cell cycle-regulating microRNA, could control the stem cell properties of adipose tissue-derived stem cells (ADSCs). First, we found that the expression level of miR-34a was increased as the cell passage number was increased. This finding, however, was inversely correlated with our finding that the overexpression of miR-34a induced the decrease of cell proliferation. In addition, miR-34a overexpression decreased the expression of various cell cycle regulators such as CDKs (-2, -4, -6) and cyclins (-E, -D), but not p21 and p53. The cell cycle analysis showed accumulation of dividing cells at S phase by miR-34a, which was reversible by co-treatment with anti-miR-34a. The potential of adipogenesis and osteogenesis of ADSCs was also decreased by miR-34a overexpression, which was recovered by co-treatment with anti-miR-34a. The surface expression of stem cell markers including CD44 was also down-regulated by miR-34a overexpression as similar to that elicited by cell cycle inhibitors. miR-34a also caused a significant decrease in mRNA expression of stem cell transcription factors as well as STAT-3 expression and phosphorylation. Cytokine profiling revealed that miR-34a significantly modulated IL-6 and -8 production, which was strongly related to cellular senescence. These data suggest the importance of miR-34a for the fate of ADSCs toward senescence rather than differentiation.


Asunto(s)
Adipogénesis/genética , Ciclo Celular/genética , Senescencia Celular/genética , MicroARNs/genética , Factor de Transcripción STAT3/metabolismo , Células Madre/citología , Tejido Adiposo/citología , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/genética , Humanos , Receptores de Hialuranos/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , MicroARNs/antagonistas & inhibidores , Oligorribonucleótidos Antisentido/farmacología , Osteogénesis/genética , Células Madre/fisiología
8.
J Biol Chem ; 286(8): 6033-9, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21156798

RESUMEN

Mammalian pre-mRNA alternative splicing mechanisms are typically studied using artificial minigenes in cultured cells, conditions that may not accurately reflect the physiological context of either the pre-mRNA or the splicing machinery. Here, we describe a strategy to investigate splicing of normal endogenous full-length pre-mRNAs under physiological conditions in live mice. This approach employs antisense vivo-morpholinos (vMOs) to mask cis-regulatory sequences or to disrupt splicing factor expression, allowing functional evaluation of splicing regulation in vivo. We applied this strategy to gain mechanistic insight into alternative splicing events involving exons 2 and 16 (E2 and E16) that control the structure and function of cytoskeletal protein 4.1R. In several mouse tissues, inclusion of E16 was substantially inhibited by interfering with a splicing enhancer mechanism using a target protector morpholino that blocked Fox2-dependent splicing enhancers in intron 16 or a splice-blocking morpholino that disrupted Fox2 expression directly. For E2, alternative 3'-splice site choice is coordinated with upstream promoter use across a long 5'-intron such that E1A splices almost exclusively to the distal acceptor (E2dis). vMOs were used to test the in vivo relevance of a deep intron element previously proposed to determine use of E2dis via a two-step intrasplicing model. Two independent vMOs designed against this intronic regulatory element inhibited intrasplicing, robustly switching E1A splicing to the proximal acceptor (E2prox). This finding strongly supports the in vivo physiological relevance of intrasplicing. vMOs represent a powerful tool for alternative splicing studies in vivo and may facilitate exploration of alternative splicing networks in vivo.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Oligorribonucleótidos Antisentido/farmacología , ARN Mensajero/biosíntesis , Empalme Alternativo/genética , Animales , Proteínas Sanguíneas/biosíntesis , Proteínas Sanguíneas/genética , Proteína p300 Asociada a E1A/biosíntesis , Proteína p300 Asociada a E1A/genética , Exones/genética , Ratones , Proteínas de Microfilamentos , ARN Mensajero/genética
9.
RNA ; 16(12): 2529-36, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20935067

RESUMEN

The Argonaute proteins play essential roles in development and cellular metabolism in many organisms, including plants, flies, worms, and mammals. Whereas in organisms such as Caenorhabditis elegans and Arabidopsis thaliana, creation of Argonaute mutant strains allowed the study of their biological functions, in mammals the application of this approach is limited by its difficulty and in the specific case of Ago2 gene, by the lethality of such mutation. Hence, in human cells, functional studies of Ago proteins relied on phenotypic suppression using small interfering RNA (siRNA) which involves Ago proteins and the RNA interference mechanism. This bears the danger of undesired or unknown interference effects which may lead to misleading results. Thus, alternative methods acting by different regulatory mechanisms would be advantageous in order to exclude unspecific effects. The knockdown may be achieved by using specific antisense oligonucleotides (asONs) which act via an RNase H-dependent mechanism, not thought to interfere with processes in which Agos are involved. Different functional observations in the use of siRNA versus asONs indicate the relevance of this assumption. We developed asONs specific for the four human Agos (hAgos) and compared their activities with those obtained by siRNA. We confirm that hAgo2 is involved in microRNA (miRNA)- and in siRNA-mediated silencing pathways, while the other hAgos play a role only in miRNA-based gene regulation. Using combinations of asONs we found that the simultaneous down-regulation of hAgo1, hAgo2, and hAgo4 led to the strongest decrease in miRNA activity, indicating a main role of these proteins.


Asunto(s)
Factor 2 Eucariótico de Iniciación/análisis , Factor 2 Eucariótico de Iniciación/genética , Oligorribonucleótidos Antisentido/farmacología , Proteínas Argonautas , Células Cultivadas , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Eficiencia , Factor 2 Eucariótico de Iniciación/antagonistas & inhibidores , Factor 2 Eucariótico de Iniciación/fisiología , Factores Eucarióticos de Iniciación/análisis , Factores Eucarióticos de Iniciación/genética , Humanos , Concentración 50 Inhibidora , Oligorribonucleótidos Antisentido/síntesis química , Interferencia de ARN/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Especificidad por Sustrato/efectos de los fármacos , Transfección
10.
Bioorg Med Chem Lett ; 22(8): 2681-3, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22460027

RESUMEN

This Letter describes the synthesis and properties of double-stranded antisense oligonucleotides connected with a pentaerythritol linker. We found that double-stranded antisense oligonucleotides with aminomethyl residues have high affinity for single-stranded DNA or RNA in buffer solutions with and without MgCl(2). Thus, these oligonucleotides would be useful as antisense oligonucleotides for targeting single-stranded RNA through triplex formation.


Asunto(s)
ADN de Cadena Simple/química , Oligorribonucleótidos Antisentido/química , ARN/química , Timidina/análogos & derivados , ADN/química , ADN de Cadena Simple/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Estabilidad de Medicamentos , Enlace de Hidrógeno , Concentración de Iones de Hidrógeno , Cloruro de Magnesio/química , Oligorribonucleótidos Antisentido/farmacología , ARN/efectos de los fármacos , Temperatura , Timidina/química
11.
Genesis ; 49(3): 117-23, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21442720

RESUMEN

Antisense oligonucleotides are commonly employed to study the roles of genes in development. Although morpholino phosphorodiamidate oligonucleotides (morpholinos) are widely used to block translation or splicing of target gene products' the usefulness of other modifications in mediating RNase-H independent inhibition of gene activity in embryos has not been investigated. In this study, we investigated the extent that fully modified 2'-O-methyl oligonucleotides (2'-OMe oligos) that can function as translation inhibiting reagents in vivo, using Xenopus and zebrafish embryos. We find that oligos against Xenopus ß-catenin, wnt11, and bmp4 and against zebrafish chordin (chd), which can efficiently and specifically generate embryonic loss-of-function phenotypes comparable with morpholino injection and other methods. These results show that fully modified 2'-OMe oligos can function as RNase-H independent antisense reagents in vertebrate embryos and can thus serve as an alternative modification to morpholinos in some cases.


Asunto(s)
Oligorribonucleótidos Antisentido/genética , Xenopus laevis/embriología , Xenopus laevis/genética , Pez Cebra/embriología , Pez Cebra/genética , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Glicoproteínas/genética , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Morfolinas , Morfolinos , Oligorribonucleótidos Antisentido/farmacología , Fenotipo , Empalme del ARN , ARN Mensajero , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Pez Cebra/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
12.
J Neurosci ; 30(43): 14513-21, 2010 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-20980609

RESUMEN

During development, neurons migrate considerable distances to reside in locations that enable their individual functional roles. Whereas migration mechanisms have been extensively studied, much less is known about how neurons remain in their ideal locations. We sought to identify factors that maintain the position of postmigratory dorsal root ganglion neurons, neural crest derivatives for which migration and final position play an important developmental role. We found that an early developing population of sensory neurons maintains the position of later born dorsal root ganglia neurons in an activity-dependent manner. Further, inhibiting or increasing the function of brain-derived neurotrophic factor induces or prevents, respectively, migration of dorsal root ganglia neurons out of the ganglion to locations where they acquire a new identity. Overall, the results demonstrate that neurotrophins mediate non-cell-autonomous maintenance of position and thereby the identity of differentiated neurons.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/fisiología , Células Receptoras Sensoriales/fisiología , Animales , Animales Modificados Genéticamente , Anticuerpos Bloqueadores/farmacología , Factor Neurotrófico Derivado del Encéfalo/antagonistas & inhibidores , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Dendritas/fisiología , Ganglios Espinales/embriología , Ganglios Espinales/crecimiento & desarrollo , Ganglios Espinales/fisiología , Inmunohistoquímica , Técnicas In Vitro , Microscopía Confocal , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Factores de Crecimiento Nervioso/fisiología , Cresta Neural/fisiología , Oligorribonucleótidos Antisentido/farmacología , Estimulación Física , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/efectos de los fármacos , Canales de Sodio/fisiología , Tetrodotoxina/farmacología , Pez Cebra
13.
J Neurosci ; 30(23): 7783-92, 2010 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-20534827

RESUMEN

The hypothalamic peptide, nesfatin-1, derived from the precursor NEFA/nucleobindin 2 (NUCB2), was recently identified as anorexigenic signal, acting in a leptin-independent manner. Yet its participation in the regulation of other biological functions gated by body energy status remains unexplored. We show herein that NUCB2/nesfatin-1 is involved in the control of female puberty. NUCB2/nesfatin mRNA and protein were detected at the hypothalamus of pubertal female rats, with prominent signals at lateral hypothalamus (LHA), paraventricular (PVN), and supraoptic (SON) nuclei. Hypothalamic NUCB2 expression raised along pubertal transition, with detectable elevations of its mRNA levels at LHA, PVN, and SON, and threefold increase of its total protein content between late-infantile and peripubertal periods. Conditions of negative energy balance, such as 48 h fasting or sustained subnutrition, decreased hypothalamic NUCB2 mRNA and/or protein levels in pubertal females. At this age, central administration of nesfatin-1 induced modest but significant elevations of circulating gonadotropins, whose magnitude was notably augmented in conditions of food deprivation. Continuous intracerebroventricular infusion of antisense morpholino oligonucleotides (as-MONs) against NUCB2 along pubertal maturation, which markedly reduced hypothalamic NUCB2 protein content, delayed vaginal opening and decreased ovarian weights and serum luteinizing hormone (LH) levels. In contrast, in adult female rats, intracerebroventricular injection of nesfatin did not stimulate LH or follicle-stimulating hormone secretion; neither did central as-MON infusion alter preovulatory gonadotropin surges, despite suppression of hypothalamic NUCB2. In sum, our data are the first to disclose the indispensable role of NUCB2/nesfatin-1 in the central networks driving puberty onset, a function that may contribute to its functional coupling to energy homeostasis.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al ADN/metabolismo , Hipotálamo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/farmacología , Neuropéptidos/farmacología , Maduración Sexual/efectos de los fármacos , Envejecimiento/efectos de los fármacos , Envejecimiento/metabolismo , Animales , Proteínas de Unión al Calcio/genética , Proteínas de Unión al ADN/genética , Femenino , Hormona Folículo Estimulante/sangre , Área Hipotalámica Lateral/metabolismo , Inyecciones Intraventriculares , Hormona Luteinizante/sangre , Proteínas del Tejido Nervioso/administración & dosificación , Proteínas del Tejido Nervioso/genética , Neuropéptidos/administración & dosificación , Neuropéptidos/metabolismo , Nucleobindinas , Oligorribonucleótidos Antisentido/administración & dosificación , Oligorribonucleótidos Antisentido/farmacología , Núcleo Hipotalámico Paraventricular/metabolismo , ARN Mensajero , Ratas , Ratas Wistar , Núcleo Supraóptico/metabolismo
14.
J Neurosci ; 30(21): 7423-33, 2010 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-20505109

RESUMEN

The chemokine SDF1 activates a cAMP-mediated signaling pathway that antagonizes retinal responses to the midline repellent slit. We show that knocking down the calmodulin-activated adenylate cyclase ADCY8 makes retinal axons insensitive to SDF1. Experiments in vivo using male and female zebrafish (Danio rerio) confirm a mutual antagonism between slit signaling and ADCY8-mediated signaling. Unexpectedly, knockdown of ADCY8 or another calmodulin-activated cyclase, ADCY1, induces ipsilateral misprojections of retinal axons that would normally cross the ventral midline. We demonstrate a cell-autonomous requirement for ADCY8 in retinal neurons for normal midline crossing. These findings are the first to show that ADCY8 is required for axonal pathfinding before axons reach their targets. They support a model in which ADCY8 is an essential component of a signaling pathway that opposes repellent signaling. Finally, they demonstrate that ADCY8 helps regulate retinal sensitivity to midline guidance cues.


Asunto(s)
Adenilil Ciclasas/fisiología , Axones/fisiología , Movimiento Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Retina/citología , Células Ganglionares de la Retina/citología , Adenilil Ciclasas/genética , Animales , Animales Modificados Genéticamente , Axones/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Trasplante de Células , Células Cultivadas , Quimiocina CXCL12/farmacología , Embrión de Pollo , Colforsina/farmacología , AMP Cíclico/metabolismo , Electroporación/métodos , Embrión no Mamífero , Femenino , Lateralidad Funcional , Proteína GAP-43/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Oligorribonucleótidos Antisentido/farmacología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Transfección , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
15.
J Biol Chem ; 285(52): 41001-9, 2010 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-20961855

RESUMEN

The mechanism that regulates embryonic liver morphogenesis remains elusive. Progranulin (PGRN) is postulated to play a critical role in regulating pathological liver growth. Nevertheless, the exact regulatory mechanism of PGRN in relation to its functional role in embryonic liver development remains to be elucidated. In our study, the knockdown of progranulin A (GrnA), an orthologue of mammalian PGRN, using antisense morpholinos resulted in impaired liver morphogenesis in zebrafish (Danio rerio). The vital role of GrnA in hepatic outgrowth and not in liver bud formation was further confirmed using whole-mount in situ hybridization markers. In addition, a GrnA deficiency was also found to be associated with the deregulation of MET-related genes in the neonatal liver using a microarray analysis. In contrast, the decrease in liver size that was observed in grnA morphants was avoided when ectopic MET expression was produced by co-injecting met mRNA and grnA morpholinos. This phenomenon suggests that GrnA might play a role in liver growth regulation via MET signaling. Furthermore, our study has shown that GrnA positively modulates hepatic MET expression both in vivo and in vitro. Therefore, our data have indicated that GrnA plays a vital role in embryonic liver morphogenesis in zebrafish. As a result, a novel link between PGRN and MET signaling is proposed.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Hígado/embriología , Organogénesis/fisiología , Proteínas Proto-Oncogénicas c-met/biosíntesis , Transducción de Señal/fisiología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intercelular/genética , Oligorribonucleótidos Antisentido/genética , Oligorribonucleótidos Antisentido/farmacología , Organogénesis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-met/genética , Transducción de Señal/efectos de los fármacos , Pez Cebra/genética , Proteínas de Pez Cebra/genética
16.
Neurobiol Dis ; 44(2): 239-47, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21784155

RESUMEN

The glycoprotein reelin is important for embryonic neuronal migration. During adulthood reelin possibly acts as a modulator of synaptic plasticity. Several studies link reduced levels of reelin messenger RNA and protein to the pathophysiology of certain neuropsychiatric disorders. However, little is known about reelin's role for behavioral and cognitive functions in vivo. Therefore, the effect of a reelin knockdown in the medial prefrontal cortex (mPFC) of Wistar rats was examined in behavioral tasks related to neuropsychiatric disorders, such as schizophrenia. Rats treated with reelin antisense phosphothioate oligonucleotides in the mPFC during puberty or adulthood were tested for prepulse inhibition (PPI) of the acoustic startle reflex, spatial working memory, object recognition, and locomotor activity. Reelin quantification in the mPFC was assessed by Western blotting. Local reelin knockdown during puberty or adulthood induced (1) a PPI deficit as well as (2) an impairment of spatial working memory and object recognition following pubertal injections. Western blot analyses showed a distinct and highly selective reelin knockdown in the rats' mPFC. These results indicate that mPFC reelin signaling plays an important role in behavioral tasks with relevance to e.g. schizophrenia. Understanding reelin's function as a neurotrophic modulator of the extracellular matrix may help to achieve new insights into the etiology of certain neuropsychiatric diseases and foster prospective treatment strategies.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/deficiencia , Moléculas de Adhesión Celular Neuronal/genética , Trastornos del Conocimiento/fisiopatología , Proteínas de la Matriz Extracelular/deficiencia , Proteínas de la Matriz Extracelular/genética , Técnicas de Silenciamiento del Gen , Trastornos del Humor/fisiopatología , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Corteza Prefrontal/fisiopatología , Serina Endopeptidasas/deficiencia , Serina Endopeptidasas/genética , Factores de Edad , Animales , Moléculas de Adhesión Celular Neuronal/antagonistas & inhibidores , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/metabolismo , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/antagonistas & inhibidores , Técnicas de Silenciamiento del Gen/métodos , Masculino , Trastornos del Humor/genética , Trastornos del Humor/metabolismo , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Oligorribonucleótidos Antisentido/farmacología , Corteza Prefrontal/crecimiento & desarrollo , Corteza Prefrontal/metabolismo , Ratas , Ratas Wistar , Proteína Reelina , Maduración Sexual/genética
17.
Birth Defects Res A Clin Mol Teratol ; 91(3): 129-41, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21308976

RESUMEN

BACKGROUND: Alcohol (ethanol) is a teratogen known to affect the developing eyes, face, and brain. Among the ocular defects in fetal alcohol spectrum disorder (FASD) are microphthalmia and optic nerve hypoplasia. Employing zebrafish as an FASD model provides an excellent system to analyze the molecular basis of prenatal ethanol exposure-induced defects because embryos can be exposed to ethanol at defined developmental stages and affected genetic pathways can be examined. We have previously shown that disruption of agrin function in zebrafish embryos produces microphthalmia and optic nerve hypoplasia. METHODS: Zebrafish embryos were exposed to varying concentrations of ethanol in the absence or presence of morpholino oligonucleotides (MOs) that disrupt agrin function. In situ hybridization was used to analyze ocular gene expression as a consequence of ethanol exposure and agrin knockdown. Morphologic analysis of zebrafish embryos was also conducted. RESULTS: Acute ethanol exposure induces diminished agrin gene expression in zebrafish eyes and, importantly, combined treatment with subthreshold levels of agrin MO and ethanol produces pronounced microphthalmia, markedly reduces agrin gene expression, and perturbs Pax6a and Mbx gene expression. Microphthalmia produced by combined agrin MO and ethanol treatment was rescued by sonic hedgehog (Shh) mRNA overexpression, suggesting that ethanol-mediated disruption of agrin expression results in disrupted Shh function. CONCLUSIONS: These studies illustrate the strong potential for using zebrafish as a model to aid in defining the molecular basis for ethanol's teratogenic effects. The results of this work suggest that agrin expression and function may be a target of ethanol exposure during embryogenesis.


Asunto(s)
Agrina/fisiología , Etanol/farmacología , Ojo/efectos de los fármacos , Ojo/embriología , Pez Cebra/embriología , Agrina/genética , Agrina/metabolismo , Animales , Animales Modificados Genéticamente , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Embrión no Mamífero , Exposición a Riesgos Ambientales , Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/fisiología , Microftalmía/inducido químicamente , Microftalmía/genética , Microftalmía/patología , Oligorribonucleótidos Antisentido/farmacología , Fenotipo , Pez Cebra/genética , Pez Cebra/metabolismo
18.
Arterioscler Thromb Vasc Biol ; 30(1): 24-30, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19834103

RESUMEN

BACKGROUND: Stearoyl-CoA desaturase 1 (SCD1) is a critical regulator of energy metabolism and inflammation. We have previously reported that inhibition of SCD1 in hyperlipidemic mice fed a saturated fatty acid (SFA)-enriched diet prevented development of the metabolic syndrome, yet surprisingly promoted severe atherosclerosis. In this study we tested whether dietary fish oil supplementation could prevent the accelerated atherosclerosis caused by SCD1 inhibition. METHODS AND RESULTS: LDLr(-/-), ApoB(100/100) mice were fed diets enriched in saturated fat or fish oil in conjunction with antisense oligonucleotide (ASO) treatment to inhibit SCD1. As previously reported, in SFA-fed mice, SCD1 inhibition dramatically protected against development of the metabolic syndrome, yet promoted atherosclerosis. In contrast, in mice fed fish oil, SCD1 inhibition did not result in augmented macrophage inflammatory response or severe atherosclerosis. In fact, the combined therapy of dietary fish oil and SCD1 ASO treatment effectively prevented both the metabolic syndrome and atherosclerosis. CONCLUSIONS: SCD1 ASO treatment in conjunction with dietary fish oil supplementation is an effective combination therapy to comprehensively combat the metabolic syndrome and atherosclerosis in mice.


Asunto(s)
Aterosclerosis/prevención & control , Grasas Insaturadas en la Dieta/farmacología , Aceites de Pescado/farmacología , Síndrome Metabólico/prevención & control , Oligorribonucleótidos Antisentido/farmacología , Estearoil-CoA Desaturasa/genética , Animales , Apolipoproteína B-100/genética , Apolipoproteína B-100/metabolismo , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/inmunología , Terapia Combinada , Ácidos Grasos/farmacología , Hígado Graso/tratamiento farmacológico , Hígado Graso/prevención & control , Hiperlipidemias/tratamiento farmacológico , Hiperlipidemias/prevención & control , Resistencia a la Insulina , Macrófagos/inmunología , Masculino , Síndrome Metabólico/dietoterapia , Síndrome Metabólico/inmunología , Ratones , Ratones Mutantes , Obesidad/tratamiento farmacológico , Obesidad/prevención & control , Receptores de LDL/genética , Receptores de LDL/metabolismo , Estearoil-CoA Desaturasa/antagonistas & inhibidores , Receptor Toll-Like 4/inmunología
19.
J Pediatr Gastroenterol Nutr ; 52(3): 339-44, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21336163

RESUMEN

BACKGROUND: zfhz1b is the causative gene for Mowat-Wilson syndrome, in which patients demonstrate developmental delay and Hirschsprung disease, as well as other anomalies. MATERIALS AND METHODS: We identified a patient with Mowat-Wilson syndrome who also developed cholestasis and histopathologic features consistent with biliary atresia, suggesting that mutations involving zfhz1b may lead to biliary developmental anomalies or injury to the biliary tract. We used the zebrafish model system to determine whether zfhx1b has a role in vertebrate biliary development. RESULTS: Using zebrafish we determined that zfhx1b was expressed in the developing liver during biliary growth and remodeling, and that morpholino antisense oligonucleotide-mediated knockdown of zfhx1b led to defects in biliary development. These findings were associated with decreased expression of vhnf1, a transcription factor known to be important in biliary development in zebrafish and in mammals. CONCLUSIONS: Our studies underscore the importance of genetic contributions in the etiology of infantile hepatobiliary disorders, including biliary atresia.


Asunto(s)
Sistema Biliar/crecimiento & desarrollo , Genes Homeobox , Enfermedad de Hirschsprung/genética , Proteínas de Homeodominio/genética , Discapacidad Intelectual/genética , Microcefalia/genética , Mutación , Proteínas Represoras/genética , Dedos de Zinc , Animales , Atresia Biliar/etiología , Atresia Biliar/genética , Atresia Biliar/metabolismo , Sistema Biliar/anomalías , Facies , Factor Nuclear 1-beta del Hepatocito/metabolismo , Enfermedad de Hirschsprung/complicaciones , Enfermedad de Hirschsprung/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Lactante , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/metabolismo , Hígado/metabolismo , Masculino , Microcefalia/complicaciones , Microcefalia/metabolismo , Oligorribonucleótidos Antisentido/farmacología , Proteínas Represoras/metabolismo , Pez Cebra , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc
20.
Dev Dyn ; 239(4): 1124-35, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20235223

RESUMEN

The N-terminus of telethonin forms a unique structure linking two titin N-termini at the Z-disc. While a specific role for the C-terminus has not been established, several studies indicate it may have a regulatory function. Using a morpholino approach in Xenopus, we show that telethonin knockdown leads to embryonic paralysis, myocyte defects, and sarcomeric disruption. These myopathic defects can be rescued by expressing full-length telethonin mRNA in morpholino background, indicating that telethonin is required for myofibrillogenesis. However, a construct missing C-terminal residues is incapable of rescuing motility or sarcomere assembly in cultured myocytes. We, therefore, tested two additional constructs: one where four C-terminal phosphorylatable residues were mutated to alanines and another where terminal residues were randomly replaced. Data from these experiments support that the telethonin C-terminus is required for assembly, but in a context-dependent manner, indicating that factors and forces present in vivo can compensate for C-terminal truncation or mutation.


Asunto(s)
Desarrollo de Músculos/genética , Proteínas Musculares/química , Proteínas Musculares/fisiología , Sarcómeros/metabolismo , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Modelos Moleculares , Desarrollo de Músculos/efectos de los fármacos , Desarrollo de Músculos/fisiología , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/fisiología , Proteínas Musculares/genética , Músculo Estriado/crecimiento & desarrollo , Músculo Estriado/metabolismo , Músculo Estriado/fisiología , Oligorribonucleótidos Antisentido/farmacología , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , Sarcómeros/efectos de los fármacos , Sarcómeros/genética , Sarcómeros/fisiología , Homología de Secuencia , Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA