Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
PLoS Genet ; 17(2): e1009318, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33600407

RESUMEN

The generation of lineage-specific gene expression programmes that alter proliferation capacity, metabolic profile and cell type-specific functions during differentiation from multipotent stem cells to specialised cell types is crucial for development. During differentiation gene expression programmes are dynamically modulated by a complex interplay between sequence-specific transcription factors, associated cofactors and epigenetic regulators. Here, we study U-shaped (Ush), a multi-zinc finger protein that maintains the multipotency of stem cell-like hemocyte progenitors during Drosophila hematopoiesis. Using genomewide approaches we reveal that Ush binds to promoters and enhancers and that it controls the expression of three gene classes that encode proteins relevant to stem cell-like functions and differentiation: cell cycle regulators, key metabolic enzymes and proteins conferring specific functions of differentiated hemocytes. We employ complementary biochemical approaches to characterise the molecular mechanisms of Ush-mediated gene regulation. We uncover distinct Ush isoforms one of which binds the Nucleosome Remodeling and Deacetylation (NuRD) complex using an evolutionary conserved peptide motif. Remarkably, the Ush/NuRD complex specifically contributes to the repression of lineage-specific genes but does not impact the expression of cell cycle regulators or metabolic genes. This reveals a mechanism that enables specific and concerted modulation of functionally related portions of a wider gene expression programme. Finally, we use genetic assays to demonstrate that Ush and NuRD regulate enhancer activity during hemocyte differentiation in vivo and that both cooperate to suppress the differentiation of lamellocytes, a highly specialised blood cell type. Our findings reveal that Ush coordinates proliferation, metabolism and cell type-specific activities by isoform-specific cooperation with an epigenetic regulator.


Asunto(s)
Ciclo Celular/genética , Proteínas de Drosophila/metabolismo , Ácidos Grasos/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Hematopoyesis/genética , Hemocitos/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Factores de Transcripción/metabolismo , Secuencias de Aminoácidos , Animales , Línea Celular , Proliferación Celular/genética , Supervivencia Celular/genética , Secuenciación de Inmunoprecipitación de Cromatina , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Elementos de Facilitación Genéticos , Ontología de Genes , Regiones Promotoras Genéticas , Isoformas de Proteínas , Interferencia de ARN , RNA-Seq , Factores de Transcripción/genética
2.
J Virol ; 94(8)2020 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-32024773

RESUMEN

Optimization of immunogen is crucial for induction of effective T-cell responses in the development of a human immunodeficiency virus (HIV) vaccine. Conventional T-cell-based vaccines have been designed to induce virus-specific CD4+ T as well as CD8+ T cells. However, it has been indicated that induction of HIV-specific CD4+ T cells, preferential targets for HIV infection, by vaccination may be detrimental and accelerate viral replication after HIV exposure. In the present study, we present a novel immunogen to selectively induce CD8+ T cells but not CD4+ T cells targeting viral antigens. The immunogen, CaV11, was constructed by tandem connection of overlapping 11-mer peptides spanning simian immunodeficiency virus (SIV) Gag capsid (CA) and Vif. Prime-boost immunization with DNA and Sendai virus (SeV) vectors expressing CaV11 efficiently induced Gag/Vif-specific CD8+ T-cell responses with inefficient Gag/Vif-specific CD4+ T-cell induction in rhesus macaques (n = 6). None of the macaques exhibited the enhancement of acute viral replication after an intravenous high-dose SIV challenge, which was observed in those immunized with DNA and SeV expressing the whole Gag protein in our previous study. Set point viral control postinfection was associated with SeV-specific CD4+ T-cell responses postimmunization, suggesting contribution of SeV-specific helper responses to effective Gag/Vif-specific CD8+ T-cell induction by vaccination. This immunogen design could be a promising method for selective induction of effective anti-HIV CD8+ T-cell responses.IMPORTANCE Induction of effective CD8+ T-cell responses is an important HIV vaccine strategy. Several promising vaccine delivery tools have been developed, and immunogen optimization is now crucial for effective T-cell induction. Conventional immunogens have been designed to induce virus-specific CD4+ T cells as well as CD8+ T cells, but induction of virus-specific CD4+ T cells that are preferential targets for HIV infection could enhance acute HIV proliferation. Here, we designed a novel immunogen to induce HIV-specific CD8+ T cells without HIV-specific CD4+ T-cell induction but with non-HIV antigen-specific CD4+ T-cell help. Our analysis in a macaque AIDS model showed that our immunogen can efficiently elicit effective CD8+ T but not CD4+ T cells targeting viral antigens, resulting in no enhancement of acute viral replication after virus exposure. This immunogen design, also applicable for other currently developed immunogens, could be a promising method for selective induction of effective anti-HIV CD8+ T-cell responses.


Asunto(s)
Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por VIH/inmunología , Vacunas contra el SIDA/inmunología , Secuencia de Aminoácidos , Animales , Modelos Animales de Enfermedad , Inmunización , Macaca mulatta , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunación , Carga Viral , Replicación Viral , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/inmunología
3.
Genesis ; 56(5): e23210, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29663653

RESUMEN

Hedgehog (Hh) pathway signaling is crucial for the maintenance of blood cell progenitors in the lymph gland hematopoietic organ present in Drosophila third instar larvae. Previous studies from our lab have likewise shown the importance of the mir-7 and bag of marbles (bam) genes in maintaining the progenitor state. Thus, we sought to investigate a possible interaction between the Hh pathway and mir-7/bam in the prohemocyte population within this hematopoietic tissue. Gain of function mir-7 was able to rescue a blood cell progenitor depletion phenotype caused by Patched (Ptc) inhibition of Hh pathway signaling in these cells. Similarly, expression of a dominant/negative version of Ptc was able to rescue the severe reduction of prohemocytes due to bam loss of function. Furthermore, we demonstrated that Suppressor of fused [Su(fu)], another known inhibitor of Hh signaling, likely serves as a translational repression target of the mir-7 miRNA. Our results suggest the mir-7/bam combination regulates the Hh signaling network through repression of Su(fu) to maintain hemocyte progenitors in the larval lymph gland.


Asunto(s)
Proteínas de Drosophila/metabolismo , MicroARNs/metabolismo , Animales , Células Sanguíneas , Diferenciación Celular/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Hedgehog/genética , Células Madre Hematopoyéticas/metabolismo , Larva/metabolismo , Ganglios Linfáticos/embriología , Ganglios Linfáticos/metabolismo , MicroARNs/genética , Receptores Inmunológicos/metabolismo , Proteínas Represoras/genética , Transducción de Señal
4.
Biochem Biophys Res Commun ; 502(3): 415-421, 2018 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-29856996

RESUMEN

Acute inflammation can cause serious tissue damage and disease in physiologically-challenged organisms. The precise mechanisms leading to these detrimental effects remain to be determined. In this study, we utilize a reproducible means to induce cellular immune activity in Drosophila larvae in response to mechanical stress. That is, forceps squeeze-administered stress induces lamellocytes, a defensive hemocyte type that normally appears in response to wasp infestation of larvae. The posterior signaling center (PSC) is a cellular microenvironment in the larval hematopoietic lymph gland that is vital for lamellocyte induction upon parasitoid attack. However, we found the PSC was not required for mechanical stress-induced lamellocyte production. In addition, we observed that mechanical injury caused a systemic expression of Unpaired3. This cytokine is both necessary and sufficient to activate the cellular immune response to the imposed stress. These findings provide new insights into the communication between injured tissues and immune system induction, using stress-challenged Drosophila larvae as a tractable model system.


Asunto(s)
Drosophila melanogaster/inmunología , Drosophila melanogaster/metabolismo , Animales , Animales Modificados Genéticamente , Microambiente Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/parasitología , Hemocitos/citología , Hemocitos/inmunología , Inmunidad Celular , Quinasas Janus/metabolismo , Larva/inmunología , Larva/metabolismo , Larva/parasitología , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Estrés Mecánico , Factores de Transcripción/metabolismo , Avispas/inmunología , Avispas/patogenicidad
5.
Development ; 142(13): 2261-7, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26041767

RESUMEN

Bag of Marbles (Bam) is known to function as a positive regulator of hematopoietic progenitor maintenance in the lymph gland blood cell-forming organ during Drosophila hematopoiesis. Here, we demonstrate a key function for Bam in cells of the lymph gland posterior signaling center (PSC), a cellular domain proven to function as a hematopoietic niche. Bam is expressed in PSC cells, and gene loss-of-function results in PSC overgrowth and disorganization, indicating that Bam plays a crucial role in controlling the proper development of the niche. It was previously shown that Insulin receptor (InR) pathway signaling is essential for proper PSC cell proliferation. We analyzed PSC cell number in lymph glands double-mutant for bam and InR pathway genes, and observed that bam genetically interacts with pathway members in the formation of a normal PSC. The elF4A protein is a translation factor downstream of InR pathway signaling, and functional knockdown of this crucial regulator rescued the bam PSC overgrowth phenotype, further supporting the cooperative function of Bam with InR pathway members. Additionally, we documented that the Retinoblastoma-family protein (Rbf), a proven regulator of cell proliferation, was present in cells of the PSC, with a bam function-dependent expression. By contrast, perturbation of Decapentaplegic or Wingless signaling failed to affect Rbf niche cell expression. Together, these findings indicate that InR pathway-Bam-Rbf functional interactions represent a newly identified means to regulate the correct size and organization of the PSC hematopoietic niche.


Asunto(s)
Tamaño de la Célula , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Células Madre Hematopoyéticas/citología , Proteína de Retinoblastoma/metabolismo , Somatomedinas/metabolismo , Nicho de Células Madre , Factores de Transcripción/metabolismo , Animales , Recuento de Células , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Epistasis Genética , Factor 4A Eucariótico de Iniciación/genética , Genes de Insecto , Células Madre Hematopoyéticas/metabolismo , Tejido Linfoide/metabolismo , Fenotipo , Células Madre Pluripotentes/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal
6.
Biochem Biophys Res Commun ; 486(4): 893-897, 2017 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-28342875

RESUMEN

In vertebrates, interaction between the nervous system and immune system is important to protect a challenged host from stress inputs from external sources. In this study, we demonstrate that sensory neurons are involved in the cellular immune response elicited by wasp infestation of Drosophila larvae. Multidendritic class IV neurons sense contacts from external stimuli and induce avoidance behaviors for host defense. Our findings show that inactivation of these sensory neurons impairs the cellular response against wasp parasitization. We also demonstrate that the nociception genes encoding the mechanosensory receptors Painless and Piezo, both expressed in class IV neurons, are essential for the normal cellular immune response to parasite challenge.


Asunto(s)
Proteínas de Drosophila/inmunología , Drosophila/inmunología , Drosophila/parasitología , Canales Iónicos/inmunología , Nociceptores/fisiología , Avispas/patogenicidad , Animales , Larva/inmunología , Larva/parasitología , Neuroinmunomodulación/inmunología
7.
Biochem Biophys Res Commun ; 453(3): 467-72, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25280996

RESUMEN

The Drosophila hematopoietic system is utilized in this study to gain novel insights into the process of growth control of the hematopoietic progenitor niche in blood development. The niche microenvironment is an essential component controlling the balance between progenitor populations and differentiated, mature blood cells and has been shown to lead to hematopoietic malignancies in humans when misregulated. MicroRNAs are one class of regulators associated with blood malignancies; however, there remains a relative paucity of information about the role of miRNAs in the niche. Here we demonstrate that bantam miRNA is endogenously active in the Drosophila hematopoietic progenitor niche, the posterior signaling center (PSC), and functions in the primary hematopoietic organ, the lymph gland, as a positive regulator of growth. Loss of bantam leads to a significant reduction in the PSC and overall lymph gland size, as well as a loss of the progenitor population and correlative premature differentiation of mature hemocytes. Interestingly, in addition to being essential for proper lymph gland development, we have determined bantam to be a novel upstream component of the insulin signaling cascade in the PSC and have unveiled dMyc as one factor central to bantam activity. These important findings identify bantam as a new hematopoietic regulator, place it in an evolutionarily conserved signaling pathway, present one way in which it is regulated, and provide a mechanism through which it facilitates cellular proliferation in the hematopoietic niche.


Asunto(s)
Drosophila/genética , Eritrocitos/citología , Células Madre Hematopoyéticas/citología , Homeostasis/fisiología , MicroARNs/fisiología , Animales
8.
Development ; 138(18): 3879-84, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21813570

RESUMEN

Bag of Marbles (Bam) is a stem cell differentiation factor in the Drosophila germ line. Here, we demonstrate that Bam has a crucial function in the lymph gland, the tissue that orchestrates the second phase of Drosophila hematopoiesis. In bam mutant larvae, depletion of hematopoietic progenitors is observed, coupled with prodigious production of differentiated hemocytes. Conversely, forced expression of Bam in the lymph gland results in expansion of prohemocytes and substantial reduction of differentiated blood cells. These findings identify Bam as a regulatory protein that promotes blood cell precursor maintenance and prevents hemocyte differentiation during larval hematopoiesis. Cell-specific knockdown of bam function via RNAi expression revealed that Bam activity is required cell-autonomously in hematopoietic progenitors for their maintenance. microRNA-7 (mir-7) mutant lymph glands present with phenotypes identical to those seen in bam-null animals and mutants double-heterozygous for bam and mir-7 reveal that the two cooperate to maintain the hematopoietic progenitor population. By contrast, analysis of yan mutant lymph glands revealed that this transcriptional regulator promotes blood cell differentiation and the loss of prohemocyte maintenance. Expression of Bam or mir-7 in hematopoietic progenitors leads to a reduction of Yan protein. Together, these results demonstrate that Bam and mir-7 antagonize the differentiation-promoting function of Yan to maintain the stem-like hematopoietic progenitor state during hematopoiesis.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila/genética , Drosophila/fisiología , Hematopoyesis/genética , Células Madre Hematopoyéticas/fisiología , Animales , Animales Modificados Genéticamente , Diferenciación Celular/genética , Proliferación Celular , Drosophila/metabolismo , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Proteínas del Ojo/fisiología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Células Germinativas/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , MicroARNs/fisiología , Análisis por Micromatrices , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas Represoras/fisiología
9.
Development ; 137(21): 3561-8, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20876645

RESUMEN

The lymph gland is a specialized organ for hematopoiesis, utilized during larval development in Drosophila. This tissue is composed of distinct cellular domains populated by blood cell progenitors (the medullary zone), niche cells that regulate the choice between progenitor quiescence and hemocyte differentiation [the posterior signaling center (PSC)], and mature blood cells of distinct lineages (the cortical zone). Cells of the PSC express the Hedgehog (Hh) signaling molecule, which instructs cells within the neighboring medullary zone to maintain a hematopoietic precursor state while preventing hemocyte differentiation. As a means to understand the regulatory mechanisms controlling Hh production, we characterized a PSC-active transcriptional enhancer that drives hh expression in supportive niche cells. Our findings indicate that a combination of positive and negative transcriptional inputs program the precise PSC expression of the instructive Hh signal. The GATA factor Serpent (Srp) is essential for hh activation in niche cells, whereas the Suppressor of Hairless [Su(H)] and U-shaped (Ush) transcriptional regulators prevent hh expression in blood cell progenitors and differentiated hemocytes. Furthermore, Srp function is required for the proper differentiation of niche cells. Phenotypic analyses also indicated that the normal activity of all three transcriptional regulators is essential for maintaining the progenitor population and preventing premature hemocyte differentiation. Together, these studies provide mechanistic insights into hh transcriptional regulation in hematopoietic progenitor niche cells, and demonstrate the requirement of the Srp, Su(H) and Ush proteins in the control of niche cell differentiation and blood cell precursor maintenance.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Drosophila , Factores de Transcripción GATA/fisiología , Proteínas Hedgehog/genética , Hematopoyesis/genética , Proteínas Represoras/fisiología , Nicho de Células Madre/metabolismo , Factores de Transcripción/fisiología , Animales , Animales Modificados Genéticamente , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proliferación Celular , Drosophila/genética , Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Drosophila/fisiología , Proteínas de Drosophila/metabolismo , Factores de Transcripción GATA/genética , Factores de Transcripción GATA/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/fisiología , Hemocitos/metabolismo , Hemocitos/fisiología , Larva/genética , Larva/metabolismo , Larva/fisiología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
Genesis ; 50(1): 41-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21809435

RESUMEN

Eater is a transmembrane protein that mediates phagocytosis in Drosophila. eater was identified in a microarray analysis of genes downregulated in S2 cells, in which Serpent had been knocked down by RNAi. The gene was shown to be expressed predominantly in plasmatocytes after embryonic development. We have extensively analyzed the transcriptional enhancer controlling eater expression with the following findings: the enhancer reproduces the plasmatocyte expression pattern of the gene as verified by anti-P1 antibody staining and a 526-basepair DNA region is active in lymph gland and hemolymph plasmatocytes. This DNA contains several GATA elements that serve as putative-binding sites for Serpent. Site-directed mutagenesis of two of these GATA sites abolishes eater expression in both lymph gland and hemolymph plasmatocytes. This suggests that Serpent regulates eater expression by binding these GATA sites, which was confirmed by gel shift analysis. These analyses allowed us to use eater-Gal4 to force plasmatocyte to lamellocyte differentiation.


Asunto(s)
Células Sanguíneas/metabolismo , Proteínas de Drosophila/genética , Drosophila/genética , Receptores de Superficie Celular/genética , Animales , Sitios de Unión/genética , Diferenciación Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Drosophila/citología , Drosophila/embriología , Proteínas de Drosophila/metabolismo , Ensayo de Cambio de Movilidad Electroforética/métodos , Elementos de Facilitación Genéticos , Factores de Transcripción GATA/genética , Factores de Transcripción GATA/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis , Hemocitos/citología , Hemocitos/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Familia de Multigenes , Mutagénesis Sitio-Dirigida , Interferencia de ARN , Receptores de Superficie Celular/metabolismo , Transcripción Genética
11.
Mol Cell Biol ; 27(11): 3962-9, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17371844

RESUMEN

Dorsal vessel morphogenesis in Drosophila melanogaster serves as a superb system with which to study the cellular and genetic bases of heart tube formation. We used a cardioblast-expressed Toll-GFP transgene to screen for additional genes involved in heart development and identified tailup as a locus essential for normal dorsal vessel formation. tailup, related to vertebrate islet1, encodes a LIM homeodomain transcription factor expressed in all cardioblasts and pericardial cells of the heart tube as well as in associated lymph gland hematopoietic organs and alary muscles that attach the dorsal vessel to the epidermis. A transcriptional enhancer regulating expression in these four cell types was identified and used as a tailup-GFP transgene with additional markers to characterize dorsal vessel defects resulting from gene mutations. Two reproducible phenotypes were observed in mutant embryos: hypoplastic heart tubes with misaligned cardioblasts and the absence of most lymph gland and pericardial cells. Conversely, a significant expansion of the lymph glands and abnormal morphology of the heart were observed when tailup was overexpressed in the mesoderm. Tailup was shown to bind to two DNA recognition sequences in the dorsal vessel enhancer of the Hand basic helix-loop-helix transcription factor gene, with one site proven to be essential for the lymph gland, pericardial cell, and Svp/Doc cardioblast expression of Hand. Together, these results establish Tailup as being a critical new transcription factor in dorsal vessel morphogenesis and lymph gland formation and place this regulator directly upstream of Hand in these developmental processes.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/anatomía & histología , Drosophila melanogaster/embriología , Corazón/embriología , Sistema Hematopoyético/embriología , Proteínas de Homeodominio/metabolismo , Morfogénesis/fisiología , Factores de Transcripción/metabolismo , Animales , Animales Modificados Genéticamente , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/fisiología , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Corazón/anatomía & histología , Sistema Hematopoyético/anatomía & histología , Proteínas de Homeodominio/genética , Fenotipo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción/genética , Transgenes
12.
Sci Rep ; 10(1): 11394, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32647227

RESUMEN

Anti-retroviral therapy (ART) can inhibit HIV proliferation but not achieve virus eradication from HIV-infected individuals. Under ART-based HIV control, virus-specific CD8+ T-cell responses are often reduced. Here, we investigated the impact of therapeutic vaccination inducing virus-specific CD8+ T-cell responses under ART on viral control in a macaque AIDS model. Twelve rhesus macaques received ART from week 12 to 32 after simian immunodeficiency virus (SIV) infection. Six of them were vaccinated with Sendai virus vectors expressing SIV Gag and Vif at weeks 26 and 32, and Gag/Vif-specific CD8+ T-cell responses were enhanced and became predominant. All macaques controlled viremia during ART but showed viremia rebound after ART cessation. Analysis of in vitro CD8+ cell ability to suppress replication of autologous lymphocytes-derived SIVs found augmentation of anti-SIV efficacy of CD8+ cells after vaccination. In the vaccinated animals, the anti-SIV efficacy of CD8+ cells at week 34 was correlated positively with Gag-specific CD8+ T-cell frequencies and inversely with rebound viral loads at week 34. These results indicate that Gag-specific CD8+ T-cell induction by therapeutic vaccination can augment anti-virus efficacy of CD8+ cells, which may be insufficient for functional cure but contribute to more stable viral control under ART.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/terapia , Antirretrovirales/farmacología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el SIDAS/administración & dosificación , Síndrome de Inmunodeficiencia Adquirida del Simio/terapia , Síndrome de Inmunodeficiencia Adquirida/inmunología , Síndrome de Inmunodeficiencia Adquirida/virología , Animales , Antirretrovirales/uso terapéutico , Linfocitos T CD8-positivos/efectos de los fármacos , Modelos Animales de Enfermedad , Productos del Gen gag/inmunología , Productos del Gen vif/inmunología , Humanos , Inmunogenicidad Vacunal , Macaca mulatta , Vacunas contra el SIDAS/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/inmunología , Carga Viral/efectos de los fármacos , Carga Viral/inmunología
13.
Genesis ; 47(11): 771-4, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19830816

RESUMEN

Based on environmental challenges or altered genetic composition, Drosophila larvae can produce up to three types of blood cells that express genetic programs essential for their distinct functions. Using transcriptional enhancers for genes expressed exclusively in plasmatocytes, crystal cells, or lamellocytes, several new hemocyte-specific enhancer-reporter transgenes were generated to facilitate the analysis of Drosophila hematopoiesis. This approach took advantage of fluorescent variants of insulated P-element reporter vectors for multilabeling cell analyses; two additional color variants were generated in these studies. These vectors were successfully used to produce transgenic fly lines that label specific hemocyte lineages with separate colors. Combining three transgene reporters allowed for the unambiguous identification of plasmatocytes, crystal cells, and lamellocytes within a complex hemocyte population. While this work focused on the hematopoietic process, these new vectors can be used to mark multiple cell types or trace complex cell lineages during any chosen aspect of Drosophila development.


Asunto(s)
Drosophila/genética , Elementos de Facilitación Genéticos , Genes Reporteros , Hematopoyesis/genética , Hemocitos/metabolismo , Transgenes , Animales
14.
Dev Biol ; 311(2): 311-23, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17936744

RESUMEN

Drosophila has emerged as an important model system to discover and analyze genes controlling hematopoiesis. One regulatory network known to control hemocyte differentiation is the Janus kinase (JAK)/Signal Transducer and Activator of Transcription (STAT) signal-transduction pathway. A constitutive activation mutation of the Janus kinase Hopscotch (hopscotch(Tumorous-lethal); hop(Tum-l)) results in a leukemia-like over-proliferation of hemocytes and copious differentiation of lamellocytes during larval stages. Here we show that the Friend of GATA (FOG) protein U-shaped (Ush) is expressed in circulating and lymph gland hemocytes, where it plays a critical role in controlling blood cell proliferation and differentiation. Our findings demonstrate that a reduction in ush function results in hematopoietic phenotypes strikingly similar to those observed in hop(Tum-l) animals. These include lymph gland hypertrophy, increased circulating hemocyte concentration, and abundant production of lamellocytes. Forced expression of N-terminal truncated versions of Ush likewise leads to larvae with severe hematopoietic anomalies. In contrast, expression of wild-type Ush results in a strong suppression of hop(Tum-l) phenotypes. Taken together, our findings demonstrate that U-shaped acts to control larval hemocyte proliferation and suppress lamellocyte differentiation, likely regulating hematopoietic events downstream of Hop kinase activity. Such functions appear to be facilitated through Ush interaction with the hematopoietic GATA factor Serpent (Srp).


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Genes Supresores de Tumor , Sistema Hematopoyético/fisiología , Hemocitos/fisiología , Quinasas Janus/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Animales , Animales Modificados Genéticamente , Diferenciación Celular/fisiología , Proliferación Celular , Proteínas de Drosophila/genética , Drosophila melanogaster/anatomía & histología , Factores de Transcripción GATA/genética , Factores de Transcripción GATA/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hemocitos/citología , Quinasas Janus/genética , Larva/citología , Larva/fisiología , Fenotipo , Factores de Transcripción/genética , Transgenes
15.
G3 (Bethesda) ; 7(2): 437-448, 2017 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-27913635

RESUMEN

A GFP expression screen has been conducted on >1000 Janelia FlyLight Project enhancer-Gal4 lines to identify transcriptional enhancers active in the larval hematopoietic system. A total of 190 enhancers associated with 87 distinct genes showed activity in cells of the third instar larval lymph gland and hemolymph. That is, gene enhancers were active in cells of the lymph gland posterior signaling center (PSC), medullary zone (MZ), and/or cortical zone (CZ), while certain of the transcriptional control regions were active in circulating hemocytes. Phenotypic analyses were undertaken on 81 of these hematopoietic-expressed genes, with nine genes characterized in detail as to gain- and loss-of-function phenotypes in larval hematopoietic tissues and blood cells. These studies demonstrated the functional requirement of the cut gene for proper PSC niche formation, the hairy, Btk29A, and E2F1 genes for blood cell progenitor production in the MZ domain, and the longitudinals lacking, dFOXO, kayak, cap-n-collar, and delilah genes for lamellocyte induction and/or differentiation in response to parasitic wasp challenge and infestation of larvae. Together, these findings contribute substantial information to our knowledge of genes expressed during the larval stage of Drosophila hematopoiesis and newly identify multiple genes required for this developmental process.


Asunto(s)
Drosophila melanogaster/genética , Elementos de Facilitación Genéticos , Hematopoyesis/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Diferenciación Celular/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/parasitología , Factor de Transcripción E2F1/genética , Factores de Transcripción Forkhead/genética , Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Hemocitos/metabolismo , Larva/genética , Larva/parasitología , Proteínas Tirosina Quinasas/genética , Transducción de Señal/genética , Avispas/patogenicidad
16.
Virus Res ; 86(1-2): 33-8, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12076827

RESUMEN

Sendai virus (SeV) is an enveloped virus with a nonsegmented negative strand RNA genome. The recovery of infectious virus from cDNA and generation of recombinant SeV carrying a foreign gene at the promoter proximal position has been demonstrated. In this study, we constructed a series of recombinant SeVs carrying a reporter human secreted alkaline phosphatase (SEAP) gene at each viral gene junction or the 5' distal end in order to measure the expression level of the foreign gene. We demonstrated that there was a gradient in the reporter gene expression level that depended on location, due to the polarity of transcription. In contrast, the growth and final titers of these recombinant viruses showed an opposite gradient to the foreign gene expression level. This suggests the potential for matching therapeutic gene expression level to individual therapy programs by changing the position of the foreign gene when SeVs are used as vectors for human gene therapy.


Asunto(s)
Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Virus Sendai/genética , Línea Celular , ADN Complementario/genética , Genes Reporteros , Terapia Genética , Genoma Viral , Recombinación Genética , Replicación Viral
18.
PLoS One ; 7(7): e41604, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22911822

RESUMEN

Hematopoiesis occurs in two phases in Drosophila, with the first completed during embryogenesis and the second accomplished during larval development. The lymph gland serves as the venue for the final hematopoietic program, with this larval tissue well-studied as to its cellular organization and genetic regulation. While the medullary zone contains stem-like hematopoietic progenitors, the posterior signaling center (PSC) functions as a niche microenvironment essential for controlling the decision between progenitor maintenance versus cellular differentiation. In this report, we utilize a PSC-specific GAL4 driver and UAS-gene RNAi strains, to selectively knockdown individual gene functions in PSC cells. We assessed the effect of abrogating the function of 820 genes as to their requirement for niche cell production and differentiation. 100 genes were shown to be essential for normal niche development, with various loci placed into sub-groups based on the functions of their encoded protein products and known genetic interactions. For members of three of these groups, we characterized loss- and gain-of-function phenotypes. Gene function knockdown of members of the BAP chromatin-remodeling complex resulted in niche cells that do not express the hedgehog (hh) gene and fail to differentiate filopodia believed important for Hh signaling from the niche to progenitors. Abrogating gene function of various members of the insulin-like growth factor and TOR signaling pathways resulted in anomalous PSC cell production, leading to a defective niche organization. Further analysis of the Pten, TSC1, and TSC2 tumor suppressor genes demonstrated their loss-of-function condition resulted in severely altered blood cell homeostasis, including the abundant production of lamellocytes, specialized hemocytes involved in innate immune responses. Together, this cell-specific RNAi knockdown survey and mutant phenotype analyses identified multiple genes and their regulatory networks required for the normal organization and function of the hematopoietic progenitor niche within the lymph gland.


Asunto(s)
Diferenciación Celular/genética , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Redes Reguladoras de Genes , Células Madre Hematopoyéticas/citología , Ganglios Linfáticos/citología , Nicho de Células Madre/genética , Animales , Biomarcadores/metabolismo , Células Sanguíneas/metabolismo , Recuento de Células , Ensamble y Desensamble de Cromatina , Dieta , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Conducta Alimentaria , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes de Insecto/genética , Células Madre Hematopoyéticas/metabolismo , Hemolinfa/citología , Hemolinfa/metabolismo , Homeostasis/genética , Larva/anatomía & histología , Larva/citología , Ganglios Linfáticos/anatomía & histología , Ganglios Linfáticos/metabolismo , Mutación/genética , Tamaño de los Órganos/genética , Especificidad de Órganos/genética , Interferencia de ARN , Transducción de Señal/genética
19.
Fly (Austin) ; 4(4): 338-43, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20798602

RESUMEN

The expression of toxic viral proteins for the purpose of eliminating distinct populations of cells, while leaving the rest of an organism unaffected, is a valuable method for analyzing development. Using the Gal4-UAS system, we employed the M2(H37A) toxic ion channel of the influenza-A virus to selectively ablate the Drosophila eye-antennal imaginal discs, hemocytes, dorsal vessel and nervous tissue, and comparatively monitored the effects of expressing the apoptosis-promoting protein Reaper in identical cell populations. In this report, we demonstrate the effectiveness of M2(H37A)-mediated ablation as a new means to selectively eliminate cells of interest during Drosophila development.


Asunto(s)
Drosophila/genética , Ingeniería Genética/métodos , Canales Iónicos/genética , Proteínas de la Matriz Viral/genética , Animales , Animales Modificados Genéticamente/crecimiento & desarrollo , Animales Modificados Genéticamente/metabolismo , Apoptosis , Drosophila/citología , Drosophila/crecimiento & desarrollo , Ojo/citología , Hemocitos/citología , Hemocitos/metabolismo , Larva/citología , Larva/genética , Larva/crecimiento & desarrollo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Orthomyxoviridae/genética , Fenotipo
20.
PLoS One ; 4(7): e6429, 2009 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-19641625

RESUMEN

Drosophila has emerged as an excellent model system in which to study cellular and genetic aspects of hematopoiesis. Under normal developmental conditions and in wild-type genetic backgrounds, Drosophila possesses two types of blood cells, crystal cells and plasmatocytes. Upon infestation by a parasitic wasp or in certain altered genetic backgrounds, a third hemocyte class called the lamellocyte becomes apparent. Herein we describe the characterization of a novel transcriptional regulatory module, a lamellocyte-active enhancer of the misshapen gene. This transcriptional control sequence appears to be inactive in all cell types of the wild-type larva, including crystal cells and plasmatocytes. However, in lamellocytes induced by wasp infestation or by particular genetic conditions, the enhancer is activated and it directs reporter GFP or DsRed expression exclusively in lamellocytes. The lamellocyte control region was delimited to a 140-bp intronic sequence that contains an essential DNA recognition element for the AP-1 transcription factor. Additionally, mutation of the kayak gene encoding the dFos subunit of AP-1 led to a strong suppression of lamellocyte production in tumorous larvae. As misshapen encodes a protein kinase within the Jun N-terminal kinase signaling pathway that functions to form an active AP-1 complex, the lamellocyte-active enhancer likely serves as a transcriptional target within a genetic auto-regulatory circuit that promotes the production of lamellocytes in immune-challenged or genetically-compromised animals.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Elementos de Facilitación Genéticos , Proteínas Serina-Treonina Quinasas/genética , Transcripción Genética , Animales , Secuencia de Bases , ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Intrones , Datos de Secuencia Molecular , Avispas
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda