Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Genomics ; 116(5): 110898, 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-39047877

RESUMEN

Using the 10x Genomics Chromium single-cell RNA sequencing (scRNA-seq) platform, we discovered unexpected heterogeneity in an established cell line developed from the midgut of the Fall armyworm, Spodoptera frugiperda, a major global pest. We analyzed the sequences of 18,794 cells and identified ten unique cellular clusters, including stem cells, enteroblasts, enterocytes and enteroendocrine cells, characterized by the expression of specific marker genes. Additionally, these studies addressed an important knowledge gap by investigating the expression of genes coding for respiratory and midgut membrane insecticide targets classified by the Insecticide Resistance Action Committee. Dual-fluorescence tagging method, fluorescence microscopy and fluorescence-activated cell sorting confirmed the expression of midgut cell type-specific genes. Stem cells were isolated from the heterogeneous population of SfMG-0617 cells. Our results, validated by KEGG and Gene Ontology analyses and supported by Monocle 3.0, advance the fields of midgut cellular biology and establish standards for scRNA-seq studies in non-model organisms.

2.
Artículo en Inglés | MEDLINE | ID: mdl-38802613

RESUMEN

The channels commonly responsible for maintaining cell resting membrane potentials are referred to as K2P (two-P-domain K+ subunit) channels. These K+ ion channels generally remain open but can be modulated by their local environment. These channels are classified based on pharmacology, pH sensitivity, mechanical stretch, and ionic permeability. Little is known about the physiological nature of these K2P channels in invertebrates. Acidic conditions depolarize neurons and muscle fibers, which may be caused by K2P channels given that one subtype can be blocked by acidic conditions. Doxapram is used clinically as a respiratory aid known to block acid-sensitive K2P channels; thus, the effects of doxapram on the muscle fibers and synaptic transmission in larval Drosophila and crawfish were monitored. A dose-dependent response was observed via depolarization of the larval Drosophila muscle and an increase in evoked synaptic transmission, but doxapram blocked the production of action potentials in the crawfish motor neuron and had a minor effect on the resting membrane potential of the crawfish muscle. This indicates that the nerve and muscle tissues in larval Drosophila and crawfish likely express different K2P channel subtypes. Since these organisms serve as physiological models for neurobiology and physiology, it would be of interest to further investigate what types of K2P channel are expressed in these tissues. (212 words).

3.
Nanomedicine ; 43: 102565, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35595014

RESUMEN

It was hypothesized that the catalyst nanoceria can increase inflammation/oxidative stress from the basal and reduce it from the elevated state. Macrophages clear nanoceria. To test the hypothesis, M0 (non-polarized), M1- (classically activated, pro-inflammatory), and M2-like (alternatively activated, regulatory phenotype) RAW 264.7 macrophages were nanoceria exposed. Inflammatory responses were quantified by IL-1ß level, arginase activity, and RT-qPCR and metabolic changes and oxidative stress by the mito and glycolysis stress tests (MST and GST). Morphology was determined by light microscopy, macrophage phenotype marker expression, and a novel three-dimensional immunohistochemical method. Nanoceria blocked IL-1ß and arginase effects, increased M0 cell OCR and GST toward the M2 phenotype and altered multiple M1- and M2-like cell endpoints toward the M0 level. M1-like cells had greater volume and less circularity/roundness. M2-like cells had greater volume than M0 macrophages. The results are overall consistent with the hypothesis.


Asunto(s)
Arginasa , Nanoestructuras , Arginasa/metabolismo , Cerio , Humanos , Inflamación , Estrés Oxidativo
4.
Dev Biol ; 395(2): 218-31, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25245869

RESUMEN

The Janus kinase (JAK) pathway is an essential, highly re-utilized developmental signaling cascade found in most metazoans. In vertebrates, the JAK intracellular cascade mediates signaling by dozens of cytokines and growth factors. In Drosophila, the Unpaired (Upd) family, encoded by three tandemly duplicated genes, is the only class of ligands associated with JAK stimulation. Unpaired has a central role in activation of JAK for most pathway functions, while Unpaired 2 regulates body size through insulin signaling. We show here that the third member of the family, unpaired 3 (upd3), overlaps upd in expression in some tissues and is essential for a subset of JAK-mediated developmental functions. First, consistent with the known requirements of JAK signaling in gametogenesis, we find that mutants of upd3 show an age-dependent impairment of fertility in both sexes. In oogenesis, graded JAK activity stimulated by Upd specifies the fates of the somatic follicle cells. As upd3 mutant females age, defects arise that can be attributed to perturbations of the terminal follicle cells, which require the highest levels of JAK activation. Therefore, in oogenesis, the activities of Upd and Upd3 both appear to quantitatively contribute to specification of those follicle cell fates. Furthermore, the sensitization of upd3 mutants to age-related decline in fertility can be used to investigate reproductive senescence. Second, loss of Upd3 during imaginal development results in defects of adult structures, including reduced eye size and abnormal wing and haltere posture. The outstretched wing and small eye phenotypes resemble classical alleles referred to as outstretched (os) mutations that have been previously ascribed to upd. However, we show that os alleles affect expression of both upd and upd3 and map to untranscribed regions, suggesting that they disrupt regulatory elements shared by both genes. Thus the upd region serves as a genetically tractable model for coordinate regulation of tandemly duplicated gene families that are commonly found in higher eukaryotes.


Asunto(s)
Envejecimiento/genética , Proteínas de Drosophila/metabolismo , Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Pleiotropía Genética/genética , Quinasas Janus/metabolismo , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Diferenciación Celular/genética , Drosophila/genética , Ojo/crecimiento & desarrollo , Femenino , Fertilidad/genética , Técnica del Anticuerpo Fluorescente , Hibridación in Situ , Mutagénesis , Folículo Ovárico/crecimiento & desarrollo , Reacción en Cadena en Tiempo Real de la Polimerasa , Alas de Animales/crecimiento & desarrollo
5.
Development ; 139(22): 4162-71, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23093424

RESUMEN

In Drosophila, ligands of the Unpaired (Upd) family activate the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway. The JAK/STAT pathway controls many developmental events, including multiple functions in the ovary. These include an early role in the germarium for specification of stalk cells and a later role in the vitellarium to pattern the follicular epithelium surrounding each cyst. In this latter role, graded JAK/STAT activation specifies three distinct anterior follicular cell fates, suggesting that Upd is a morphogen in this system. Consistent with the JAK/STAT activation pattern in the vitellarium, Upd forms a concentration gradient on the apical surface of the follicular epithelium with a peak at its source, the polar cells. Like many morphogens, signaling and distribution of Upd are regulated by the heparan sulfate proteoglycans (HSPGs) Dally and Dally-like. Mutations in these glypican genes and in heparan sulfate biosynthetic genes result in disruption of JAK/STAT signaling, loss or abnormal formation of the stalk and significant reduction in the accumulation of extracellular Upd. Conversely, forced expression of Dally causes ectopic accumulation of Upd in follicular cells. Furthermore, biochemical studies reveal that Upd and Dally bind each other on the surface of the cell membrane. Our findings demonstrate that Drosophila glypicans regulate formation of the follicular gradient of the Upd morphogen, Upd. Furthermore, we establish the follicular epithelium as a new model for morphogen signaling in complex organ development.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/embriología , Glipicanos/metabolismo , Quinasas Janus/metabolismo , Oogénesis , Factores de Transcripción STAT/metabolismo , Factores de Transcripción/metabolismo , Animales , Tipificación del Cuerpo , Comunicación Celular , Diferenciación Celular , Membrana Celular/metabolismo , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Células Epiteliales , Regulación del Desarrollo de la Expresión Génica , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Complejos Multiproteicos/metabolismo , Proteoglicanos/genética , Proteoglicanos/metabolismo , Transducción de Señal , Sulfotransferasas/genética , Sulfotransferasas/metabolismo
6.
Methods ; 68(1): 160-72, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24685392

RESUMEN

The Janus Kinase/Signal Transducers and Activators of Transcription (JAK/STAT) signaling pathway is one of a limited number of signaling cascades that is extensively utilized for many developmental and homeostatic functions. The JAK/STAT pathway is evolutionarily conserved from insects to mammals, with homologous transduction machinery in each. Yet the mammalian pathway is composed of multiple members for each family of proteins, while flies have only a single homologue of most pathway components. This simplicity and the abundance of genetic, biochemical, and developmental tools make Drosophila an attractive model to investigate this important signaling pathway. This review provides a basic description of the Drosophila JAK/STAT cascade and summarizes currently available reagents and tools to study and manipulate the pathway.


Asunto(s)
Quinasas Janus/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal/genética , Animales , Drosophila/genética , Drosophila/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Quinasas Janus/genética , Biología Molecular/métodos , Factores de Transcripción STAT/genética
7.
Artículo en Inglés | MEDLINE | ID: mdl-39154976

RESUMEN

The gram-negative toxin lipopolysaccharides (LPS) are known to trigger inflammatory cytokines in mammals, which can result in pathological responses. Upon treatment of bacterial sepsis with antibiotics, the lysing bacteria can present a surge in LPS, inducing a cytokine storm. However, LPS can also have direct cellular effects, including transient rapid hyperpolarizing of the membrane potential, blocking glutamate receptors and even promoting release of glutamate. The detailed mechanism of action for these immediate responses is still unresolved. In addressing the membrane hyperpolarization, voltage gated K+ channel blockers 4-aminopyridine (4-AP, 3 mM), quinidine hydrochloride monohydrate (0.1 mM) and tetraethylammonium (TEA, 20 mM) were examined along with RNAi knockdowns of potential calcium activated K+ channels. The immediate responses of LPS were not blocked. Even in the presence of glutamate, the membrane still hyperpolarizes with LPS. When the driving gradient for the ionotropic glutamate receptors is enhanced during hyperpolarization, spontaneous quantal responses are dampened in amplitude. Thus, glutamate receptors are blocked, and the mechanism of hyperpolarization remains unresolved. The larval Drosophila glutamatergic neuromuscular junction is used as a model synaptic preparation to address the direct rapid actions by LPS.


Asunto(s)
Lipopolisacáridos , Potenciales de la Membrana , Animales , Lipopolisacáridos/farmacología , Potenciales de la Membrana/efectos de los fármacos , Canales de Potasio Calcio-Activados/metabolismo , Unión Neuromuscular/efectos de los fármacos , Unión Neuromuscular/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Drosophila melanogaster , Ácido Glutámico/metabolismo , Ácido Glutámico/farmacología , Membrana Celular/metabolismo , Membrana Celular/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Larva/efectos de los fármacos , Larva/metabolismo
8.
Proc Natl Acad Sci U S A ; 107(19): 8557-62, 2010 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-20410462

RESUMEN

Cytochrome P450-mediated detoxification is one of the most important mechanisms involved in insecticide resistance. However, the molecular basis of this mechanism and the physiological functions of P450s associated with insecticide resistance remain largely unknown. Here, we exploited the functional genomics and reverse genetic approaches to identify and characterize a P450 gene responsible for the majority of deltamethrin resistance observed in the QTC279 strain of Tribolium castaneum. We used recently completed whole-genome sequence of T. castaneum to prepare custom microarrays and identified a P450 gene, CYP6BQ9, which showed more than a 200-fold higher expression in the deltamethrin-resistant QTC279 strain when compared with its expression in the deltamethrin-susceptible Lab-S strain. Functional studies using both double-strand RNA (dsRNA)-mediated knockdown in the expression of CYP6BQ9 and transgenic expression of CYP6BQ9 in Drosophila melanogaster showed that CYP6BQ9 confers deltamethrin resistance. Furthermore, CYP6BQ9 enzyme expressed in baculovirus metabolizes deltamethrin to 4-hydroxy deltamethrin. Strikingly, we also found that unlike many P450 genes involved in insecticide resistance that were reported previously, CYP6BQ9 is predominantly expressed in the brain, a part of the central nervous system (CNS) containing voltage-gated sodium channels targeted by deltamethrin. Taken together, the current studies on the brain-specific insect P450 involved in deltamethrin resistance shed new light on the understanding of the molecular basis and evolution of insecticide resistance.


Asunto(s)
Encéfalo/enzimología , Sistema Enzimático del Citocromo P-450/metabolismo , Resistencia a los Insecticidas/efectos de los fármacos , Nitrilos/farmacología , Piretrinas/farmacología , Tribolium/efectos de los fármacos , Tribolium/enzimología , Animales , Animales Modificados Genéticamente , Encéfalo/efectos de los fármacos , Sistema Enzimático del Citocromo P-450/genética , Drosophila melanogaster/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Genes de Insecto/genética , Resistencia a los Insecticidas/genética , Datos de Secuencia Molecular , Especificidad de Órganos/efectos de los fármacos , Especificidad de Órganos/genética , Piretrinas/metabolismo , Interferencia de ARN/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Tribolium/genética
9.
bioRxiv ; 2023 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-36798317

RESUMEN

Apolipoprotein E4 (APOE4) is the strongest risk allele associated with the development of late onset Alzheimer's disease (AD). Across the CNS, astrocytes are the predominant expressor of APOE while also being critical mediators of neuroinflammation and cerebral metabolism. APOE4 has been consistently linked with dysfunctional inflammation and metabolic processes, yet insights into the molecular constituents driving these responses remain unclear. Utilizing complementary approaches across humanized APOE mice and isogenic human iPSC astrocytes, we demonstrate that ApoE4 alters the astrocyte immunometabolic response to pro-inflammatory stimuli. Our findings show that ApoE4-expressing astrocytes acquire distinct transcriptional repertoires at single-cell and spatially-resolved domains, which are driven in-part by preferential utilization of the cRel transcription factor. Further, inhibiting cRel translocation in ApoE4 astrocytes abrogates inflammatory-induced glycolytic shifts and in tandem mitigates production of multiple pro-inflammatory cytokines. Altogether, our findings elucidate novel cellular underpinnings by which ApoE4 drives maladaptive immunometabolic responses of astrocytes.

10.
Function (Oxf) ; 3(3): zqac015, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35434632

RESUMEN

Aging is accompanied by reduced remodeling of skeletal muscle extracellular matrix (ECM), which is exacerbated during recovery following periods of disuse atrophy. Mechanotherapy has been shown to promote ECM remodeling through immunomodulation in adult muscle recovery, but not during the aged recovery from disuse. In order to determine if mechanotherapy promotes ECM remodeling in aged muscle, we performed single cell RNA sequencing (scRNA-seq) of all mononucleated cells in adult and aged rat gastrocnemius muscle recovering from disuse, with (REM) and without mechanotherapy (RE). We show that fibroadipogenic progenitor cells (FAPs) in aged RE muscle are highly enriched in chemotaxis genes (Csf1), but absent in ECM remodeling genes compared to adult RE muscle (Col1a1). Receptor-ligand (RL) network analysis of all mononucleated cell populations in aged RE muscle identified chemotaxis-enriched gene expression in numerous stromal cell populations (FAPs, endothelial cells, pericytes), despite reduced enrichment of genes related to phagocytic activity in myeloid cell populations (macrophages, monocytes, antigen presenting cells). Following mechanotherapy, aged REM mononuclear cell gene expression resembled adult RE muscle as evidenced by RL network analyses and KEGG pathway activity scoring. To validate our transcriptional findings, ECM turnover was measured in an independent cohort of animals using in vivo isotope tracing of intramuscular collagen and histological scoring of the ECM, which confirmed mechanotherapy-mediated ECM remodeling in aged RE muscle. Our results highlight age-related cellular mechanisms underpinning the impairment to complete recovery from disuse, and also promote mechanotherapy as an intervention to enhance ECM turnover in aged muscle recovering from disuse.


Asunto(s)
Células Endoteliales , Trastornos Musculares Atróficos , Ratas , Animales , Músculo Esquelético/metabolismo , Trastornos Musculares Atróficos/metabolismo , Macrófagos , Matriz Extracelular
11.
Insects ; 12(1)2021 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-33418937

RESUMEN

The transient receptor potential (TrpA-ankyrin) receptor has been linked to pathological conditions in cardiac function in mammals. To better understand the function of the TrpA1 in regulation of the heart, a Drosophila melanogaster model was used to express TrpA1 in heart and body wall muscles. Heartbeat of in intact larvae as well as hearts in situ, devoid of hormonal and neural input, indicate that strong over-expression of TrpA1 in larvae at 30 or 37 °C stopped the heart from beating, but in a diastolic state. Cardiac function recovered upon cooling after short exposure to high temperature. Parental control larvae (UAS-TrpA1) increased heart rate transiently at 30 and 37 °C but slowed at 37 °C within 3 min for in-situ preparations, while in-vivo larvae maintained a constant heart rate. The in-situ preparations maintained an elevated rate at 30 °C. The heartbeat in the TrpA1-expressing strains could not be revived at 37 °C with serotonin. Thus, TrpA1 activation may have allowed enough Ca2+ influx to activate K(Ca) channels into a form of diastolic stasis. TrpA1 activation in body wall muscle confirmed a depolarization of membrane. In contrast, blowfly Phaenicia sericata larvae increased heartbeat at 30 and 37 °C, demonstrating greater cardiac thermotolerance.

12.
Curr Biol ; 17(9): R328-30, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17470347

RESUMEN

Sex is determined in Drosophila by the activity of the Sex-lethal master regulator. Activity of Sex-lethal is initiated early in females by chromosome-counting transcription factors, then reinforced by signaling through the Janus kinase pathway.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/fisiología , Regulación de la Expresión Génica/fisiología , Procesos de Determinación del Sexo , Transducción de Señal/fisiología , Empalme Alternativo/fisiología , Animales , Femenino , Quinasas Janus/metabolismo , Factores de Transcripción/metabolismo
13.
Dev Cell ; 4(2): 167-77, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12586061

RESUMEN

The Drosophila egg develops through closely coordinated activities of associated germline and somatic cells. An essential aspect of egg development is the differentiation of the somatic follicle cells into several distinct subpopulations with specific functions. Here we demonstrate that the graded activity of the Janus kinase (JAK) pathway, stimulated by the Unpaired ligand, patterns the anterior-posterior axis of the follicular epithelium. Different levels of JAK activity instruct adoption of distinct anterior cell fates. Further, the coordinated activities of the JAK/STAT and epidermal growth factor receptor (EGFR) pathways are required to specify the posterior terminal cell fate. We propose that Upd secreted from the polar cells may act as a morphogen to stimulate A/P-derived follicular fates through JAK pathway activation.


Asunto(s)
Drosophila melanogaster/embriología , Embrión no Mamífero/fisiología , Células Epiteliales/citología , Oogénesis/fisiología , Folículo Ovárico/fisiología , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción , Animales , Animales Modificados Genéticamente , Diferenciación Celular , Polaridad Celular , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Activación Enzimática , Receptores ErbB/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Glicoproteínas/metabolismo , Janus Quinasa 1 , Mutación , Folículo Ovárico/citología , Fenotipo , Factor de Transcripción STAT3 , Transactivadores/metabolismo
14.
Biol Open ; 8(11)2019 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-31704693

RESUMEN

Rapamycin and other mTOR inhibitors are being heralded as possible treatments for many human ailments. It is currently being utilized clinically as an immunomodulator after transplantation procedures and as a treatment for certain forms of cancer, but it has numerous potential clinical indications. Some studies have shown profound effects on life cycle and muscle physiology, but these issues have not been addressed in an organism undergoing developmental processes. This paper fills this void by examining the effect of mTOR inhibition by rapamycin on several different qualities of larval Drosophila Various dosages of the compound were fed to second instar larvae. These larvae were monitored for pupae formation to elucidate possible life cycle effects, and a delay to pupation was quantified. Behavioral deficits were documented in rapamycin-treated larvae. Electrophysiological measurements were taken to discern changes in muscle physiology and synaptic signaling (i.e. resting membrane potential, amplitude of excitatory post-synaptic potentials, synaptic facilitation). Pupation delay and effects on behavior that are likely due to synaptic alterations within the central nervous system were discovered in rapamycin-fed larvae. These results allow for several conclusions as to how mTOR inhibition by rapamycin affects a developing organism. This could eventually allow for a more informed decision when using rapamycin and other mTOR inhibitors to treat human diseases, especially in children and adolescents, to account for known side effects.

15.
Biochim Biophys Acta ; 1576(3): 287-97, 2002 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-12084575

RESUMEN

The Drosophila crooked neck (crn) gene is essential for embryogenesis and has been implicated in cell cycle progression and in pre-mRNA splicing although a direct role in either process has not been established. Here we report isolation of the human crooked neck homolog, HCRN, and provide evidence for its function in splicing. HCRN encodes an unusual protein composed largely of tetratricopeptide repeat (TPR) elements. The crooked neck protein co-localizes with the SR and Sm protein splicing factors in discrete subnuclear domains implicated in snRNP biogenesis. In vitro assembly experiments show that an 83 kDa hcrn isoform is stably recruited to splicing complexes coincident with the addition of the U4/U6.U5 tri-snRNP particle. Crooked neck activity appears essential as extracts depleted of hcrn fail to splice pre-mRNA. These and related data support the view that crooked neck is a phylogenetically conserved pre-mRNA splicing factor.


Asunto(s)
Drosophila melanogaster/genética , Proteínas/genética , Proteínas/metabolismo , Empalme del ARN , Empalmosomas/metabolismo , Secuencia de Aminoácidos , Animales , Fraccionamiento Celular , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas Fúngicas/genética , Células HeLa , Humanos , Datos de Secuencia Molecular , Proteínas Nucleares , Proteínas/química , Secuencias Repetitivas de Ácidos Nucleicos , Alineación de Secuencia , Empalmosomas/genética , Distribución Tisular
16.
BMC Cell Biol ; 5(1): 38, 2004 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15488148

RESUMEN

BACKGROUND: The Janus kinase (JAK) cascade is an essential and well-conserved pathway required to transduce signals for a variety of ligands in both vertebrates and invertebrates. While activation of the pathway is essential to many processes, mutations from mammals and Drosophila demonstrate that regulation is also critical. The SOCS (Suppressor Of Cytokine Signaling) proteins in mammals are regulators of the JAK pathway that participate in a negative feedback loop, as they are transcriptionally activated by JAK signaling. Examination of one Drosophila SOCS homologue, Socs36E, demonstrated that its expression is responsive to JAK pathway activity and it is capable of downregulating JAK signaling, similar to the well characterized mammalian SOCS. RESULTS: Based on sequence analysis of the Drosophila genome, there are three identifiable SOCS homologues in flies. All three are most similar to mammalian SOCS that have not been extensively characterized: Socs36E is most similar to mammalian SOCS5, while Socs44A and Socs16D are most similar to mammalian SOCS6 and 7. Although Socs44A is capable of repressing JAK activity in some tissues, its expression is not regulated by the pathway. Furthermore, Socs44A can enhance the activity of the EGFR/MAPK signaling cascade, in contrast to Socs36E. CONCLUSIONS: Two Drosophila SOCS proteins have some overlapping and some distinct capabilities. While Socs36E behaves similarly to the canonical vertebrate SOCS, Socs44A is not part of a JAK pathway negative feedback loop. Nonetheless, both SOCS regulate JAK and EGFR signaling pathways, albeit differently. The non-canonical properties of Socs44A may be representative of the class of less characterized vertebrate SOCS with which it shares greatest similarity.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila melanogaster/genética , Receptores ErbB/fisiología , Genoma , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Quinasas/fisiología , Proteínas Tirosina Quinasas/metabolismo , Receptores de Péptidos de Invertebrados/fisiología , Proteínas Represoras/fisiología , Transducción de Señal/fisiología , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Janus Quinasa 1 , Janus Quinasa 2 , Janus Quinasa 3 , Datos de Secuencia Molecular , Oogénesis/genética , Ovario/química , Ovario/enzimología , Ovario/metabolismo , Proteínas Tirosina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Represoras/genética , Proteínas Supresoras de la Señalización de Citocinas , Regulación hacia Arriba/fisiología , Alas de Animales/embriología , Alas de Animales/enzimología
17.
Mol Biol Cell ; 22(17): 3231-41, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21737674

RESUMEN

Ras-related small GTP-binding proteins control a wide range of cellular processes by regulating a variety of effector pathways, including prominent roles in the control of mitogen-activated protein kinase (MAPK) cascades. Although the regulatory role(s) for many Ras family GTPases are well established, the physiological function for the Rit/Rin subfamily has been lacking. Here, using both knockout mice and Drosophila models, we demonstrate an evolutionarily conserved role for Rit subfamily GTPases (mammalian Rit and Rin, and the Drosophila RIC homologue) in governing survival in response to oxidative stress. Primary embryonic fibroblasts derived from Rit knockout mice display increased apoptosis and selective disruption of MAPK signaling following reactive oxygen species (ROS) exposure but not in response to endoplasmic reticulum stress or DNA damage. These deficits include a reduction in ROS-mediated stimulation of a p38-MK2-HSP27 signaling cascade that controls Akt activation, directing Bad phosphorylation to promote cell survival. Furthermore, D-RIC null flies display increased susceptibility to environmental stresses and reduced stress-dependent p38 signaling, extending the Rit-p38 survival pathway to Drosophila. Together, our studies establish the Rit GTPases as critical regulators of an evolutionarily conserved, p38 MAPK-dependent signaling cascade that functions as an important survival mechanism for cells in response to oxidative stress.


Asunto(s)
Secuencia Conservada , Evolución Molecular , Sistema de Señalización de MAP Quinasas , Estrés Oxidativo , Proteínas ras/metabolismo , Animales , Supervivencia Celular , Células Cultivadas , Drosophila/genética , Drosophila/fisiología , Estrés del Retículo Endoplásmico , Activación Enzimática , Fibroblastos/fisiología , Técnicas de Inactivación de Genes , Peróxido de Hidrógeno/farmacología , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Oxidantes/farmacología , Fosforilación , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos , Proteínas ras/genética
19.
Semin Cell Dev Biol ; 17(4): 518-33, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16890463

RESUMEN

Stem cells are self-renewing multipotent cells essential for development or homeostasis of many tissues. Stem cell populations can be found in most multicellular plants and animals. The mechanisms by which these populations are maintained are diverse, utilizing both intrinsic and extrinsic factors to regulate cell division and differentiation. The genetic tools of the fruitfly, Drosophila melanogaster, have permitted detailed characterization of two stem cell populations. In this review, we will examine these contrasting stem cell model systems from Drosophila and their relevance to stem cell populations in other organisms.


Asunto(s)
División Celular/fisiología , Drosophila/citología , Células Madre/fisiología , Animales , Sistema Nervioso Central/citología , Gónadas/citología , Modelos Biológicos
20.
Synapse ; 57(3): 132-47, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15945059

RESUMEN

Evoked neurotransmitter release at the Drosophila neuromuscular junction (NMJ) is regulated by the amount of calcium influx at the presynaptic nerve terminal, as for most chemical synapses. Calcium entry occurs via voltage-gated calcium channels. The temperature-sensitive Drosophila mutant, cac(TS2), has a reduced amount of calcium entry during evoked stimulation. We have used this mutation to examine homeostatic regulatory mechanisms during development of the NMJ on muscle 6 within the developing larva. The amplitude of the excitatory postsynaptic potentials are reduced for both the Ib and Is motor neurons in 3rd instar larvae which have been raised at 33 degrees C from the 1st instar stage. Larvae raised at 25 degrees C and larvae pulsed at 33 degrees C from the late 2nd instar for various lengths of time show a reduced synaptic efficacy as a 3rd instar. The results indicate that the nerve terminal cannot fully compensate physiologically in the regulation of synaptic transmission during larval life for a reduced amount of evoked calcium entry. Morphological comparisons of Ib and Is terminals in relation to length and numbers of varicosities are significantly reduced in cac(TS2), which also suggests a lack in homeostatic ability. These findings are relevant since many deficits in synaptic transmission in various systems are compensated for either physiologically or structural over development, but not in this case for reduced calcium entry during evoked transmission.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Unión Neuromuscular/fisiología , Terminales Presinápticos/fisiología , Factores de Edad , Animales , Animales Modificados Genéticamente , Conducta Animal , Composición Corporal/genética , Canales de Calcio/genética , Proteínas de Drosophila/genética , Estimulación Eléctrica/métodos , Potenciales Postsinápticos Excitadores/fisiología , Conducta Alimentaria/fisiología , Peroxidasa de Rábano Silvestre/metabolismo , Larva , Microscopía Confocal/métodos , Actividad Motora/genética , Unión Neuromuscular/efectos de la radiación , Técnicas de Placa-Clamp/métodos , Terminales Presinápticos/efectos de la radiación , Estadísticas no Paramétricas , Sensación Térmica/genética , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA