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








Base de dados
Intervalo de ano de publicação
1.
Antonie Van Leeuwenhoek ; 116(8): 829-843, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37243862

RESUMO

The biofilm formation by bacteria is a complex process that is strongly mediated by various genetic and environmental factors. Biofilms contribute to disease infestation, especially in chronic infections. It is, therefore important to understand the factors affecting biofilm formation. This study reports the role of a functional amyloid curli in biofilm formation at various abiotic surfaces, including medical devices, by an environmental isolate of Enterobacter cloacae (SBP-8) which has been known for its pathogenic potential. A knockout mutant of csgA, the gene encoding the major structural unit of curli, was created to study the effect of curli on biofilm formation by E. cloacae SBP-8. Our findings confirm the production of curli at 25 °C and 37 °C in the wild-type strain. We further investigated the role of curli in the attachment of E. cloacae SBP-8 to glass, enteral feeding tube, and foley latex catheter. Contrary to the previous studies reporting the curli production below 30 °C in the majority of biofilm-forming bacterial species, we observed its production in E. cloacae SBP-8 at 37 °C. The formation of more intense biofilm in wild-type strain on various surfaces compared to curli-deficient strain (ΔcsgA) at both 25 °C and 37 °C suggested a prominent role of curli in biofilm formation. Further, electron and confocal microscopy studies demonstrated the formation of diffused monolayers of microbial cells on the abiotic surfaces by ΔcsgA strain as compared to the thick biofilm by respective wild-type strain, indicating the involvement of curli in biofilm formation by E. cloacae SBP-8. Overall, our findings provide insight into biofilm formation mediated by curli in E. cloacae SBP-8. Further, we show that it can be expressed at a physiological temperature on all surfaces, thereby indicating the potential role of curli in pathogenesis.


Assuntos
Enterobacter cloacae , Proteínas de Escherichia coli , Enterobacter cloacae/genética , Biofilmes , Proteínas Amiloidogênicas , Fímbrias Bacterianas/genética , Proteínas de Escherichia coli/genética , Proteínas de Bactérias/genética
3.
Front Cell Neurosci ; 17: 1295805, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38239290

RESUMO

One of the hallmarks of Parkinson's Disease (PD) is aggregation of incorrectly folded α-synuclein (SNCA) protein resulting in selective death of dopaminergic neurons. Another form of PD is characterized by the loss-of-function of an E3-ubiquitin ligase, parkin. Mutations in SNCA and parkin result in impaired mitochondrial morphology, causing loss of dopaminergic neurons. Despite extensive research on the individual effects of SNCA and parkin, their interactions in dopaminergic neurons remain understudied. Here we employ Drosophila model to study the effect of collective overexpression of SNCA along with the downregulation of parkin in the dopaminergic neurons of the posterior brain. We found that overexpression of SNCA along with downregulation of parkin causes a reduction in the number of dopaminergic neuronal clusters in the posterior region of the adult brain, which is manifested as progressive locomotor dysfunction. Overexpression of SNCA and downregulation of parkin collectively results in altered mitochondrial morphology in a cluster-specific manner, only in a subset of dopaminergic neurons of the brain. Further, we found that SNCA overexpression causes transcriptional downregulation of parkin. However, this downregulation is not further enhanced upon collective SNCA overexpression and parkin downregulation. This suggests that the interactions of SNCA and parkin may not be additive. Our study thus provides insights into a potential link between α-synuclein and parkin interactions. These interactions result in altered mitochondrial morphology in a cluster-specific manner for dopaminergic neurons over a time, thus unraveling the molecular interactions involved in the etiology of Parkinson's Disease.

4.
Hum Nutr Metab ; 31: 200179, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38620788

RESUMO

The vulnerability of human health is amplified in recent times with global increase in non-communicable diseases (due to lifestyle changes and environmental insults) and infectious diseases (caused by newer pathogens and drug-resistance strains). Clinical management of diseases is further complicated by disease severity caused by other comorbid factors. Drug-based therapy may not be the sole approach, particularly in scenarios like the COVID-19 pandemic, where there is no specific drug against SARS-CoV-2. Nutritional interventions are significant in armouring human populations in disease prevention, and as adjunctive therapy for disease alleviation. Amidst ongoing clinical trials to determine the efficacy of Vit. D against infections and associated complications, this review examines the pleiotropic benefits of nutritional adequacy of vitamin D (Vit. D) in combating viral infections (COVID-19), its severity and complications due to co-morbidities (obesity, diabetes, stroke and Kawasaki disease), based on research findings and clinical studies. Supplements of Vit. D in combination with other nutrients, and drugs, are suggested as promising preventive-health and adjunct-treatment strategies in the clinical management of viral infections with metabolic comorbidities.

5.
Front Microbiol ; 13: 877060, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35865928

RESUMO

Bacterial biofilms are clinically admissible and illustrate an influential role in infections, particularly those related to the implant of medical devices. The characterization of biofilms is important to understand the etiology of the diseases. Enterobacter cloacae are known for causing infections by forming biofilms on various abiotic surfaces, such as medical devices. However, a detailed characterization in terms of morphology and the molecular composition of the formed biofilms by this bacterium is sparse. The present study provides insights into the biofilm formation of E. cloacae SBP-8, an environmental isolate, on various surfaces. We performed assays to understand the biofilm-forming capability of the SBP-8 strain and characterized the adhering potential of the bacteria on the surface of different medical devices (foley latex catheter, enteral feeding tube, and glass) at different temperatures. We found that medical devices exhibited strong colonization by E. cloacae SBP-8. Using field emission-scanning electron microscopy (FE-SEM) studies, we characterized the biofilms as a function of time. It indicated stronger biofilm formation in terms of cellular density and EPS production on the surfaces. Further, we characterized the biofilm employing surface-enhanced Raman spectroscopy (SERS) and identified the vast heterogenic nature of the biofilm-forming molecules. Interestingly, we also found that this heterogeneity varies from the initial stages of biofilm formation until the maturation and dispersion. Our studies provide insights into biofilm composition over a period of time, which might aid in understanding the biofilm dispersion phases, to enhance the presently available treatment strategies.

6.
Animal Model Exp Med ; 5(1): 27-37, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35229999

RESUMO

Alzheimer's disease and Parkinson's disease are two of the most prevalent and disabling neurodegenerative diseases globally. Both are proteinopathic conditions and while occasionally inherited, are largely sporadic in nature. Although the advances in our understanding of the two have been significant, they are far from complete and neither diagnosis nor the current practices in treatment and rehabilitation is adequately helpful. Animal models have historically found application as testing beds for novel therapeutics and continue to be valuable aids in pharmacological research. This review chronicles the development of those models in the context of Alzheimer's and Parkinson's disease, and highlights the shifting paradigms in studying two human-specific conditions in non-human organisms.


Assuntos
Doença de Alzheimer , Doença de Parkinson , Doença de Alzheimer/diagnóstico , Animais , Modelos Animais , Doença de Parkinson/terapia
7.
Sci Rep ; 11(1): 2645, 2021 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-33514791

RESUMO

The initiator caspase Dronc is the only CARD-domain containing caspase in Drosophila and is essential for apoptosis. Here, we report that homozygous dronc mutant adult animals are short-lived due to the presence of a poorly developed, defective and leaky intestine. Interestingly, this mutant phenotype can be significantly rescued by enteroblast-specific expression of dronc+ in dronc mutant animals, suggesting that proper Dronc function specifically in enteroblasts, one of four cell types in the intestine, is critical for normal development of the intestine. Furthermore, enteroblast-specific knockdown of dronc in adult intestines triggers hyperplasia and differentiation defects. These enteroblast-specific functions of Dronc do not require the apoptotic pathway and thus occur in a non-apoptotic manner. In summary, we demonstrate that an apoptotic initiator caspase has a very critical non-apoptotic function for normal development and for the control of the cell lineage in the adult midgut and therefore for proper physiology and homeostasis.


Assuntos
Caspases/biossíntese , Proteínas de Drosophila/biossíntese , Regulação da Expressão Gênica , Mucosa Intestinal/metabolismo , Mutação , Animais , Apoptose , Caspases/genética , Proteínas de Drosophila/genética , Drosophila melanogaster
8.
Genesis ; 58(10-11): e23395, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32990387

RESUMO

During organogenesis, cell proliferation is followed by the differentiation of specific cell types to form an organ. Any aberration in differentiation can result in developmental defects, which can result in a partial to a near-complete loss of an organ. We employ the Drosophila eye model to understand the genetic and molecular mechanisms involved in the process of differentiation. In a forward genetic screen, we identified, cullin-4 (cul-4), which encodes an E3 ubiquitin ligase, to play an important role in retinal differentiation. During development, cul-4 is known to be involved in protein degradation, regulation of genomic stability, and regulation of cell cycle. Previously, we have reported that cul-4 regulates cell death during eye development by downregulating Wingless (Wg)/Wnt signaling pathway. We found that loss-of-function of cul-4 results in a reduced eye phenotype, which can be due to onset of cell death. However, we found that loss-of-function of cul-4 also affects retinal development by downregulating retinal determination (RD) gene expression. Early markers of retinal differentiation are dysregulated in cul-4 loss of function conditions, indicating that cul-4 is necessary for differentiation. Furthermore, loss-of-function of cul-4 ectopically induces expression of negative regulators of eye development like Wg and Homothorax (Hth). During eye development, Wg is known to block the progression of a synchronous wave of differentiation referred to as Morphogenetic furrow (MF). In cul-4 loss-of-function background, expression of dpp-lacZ, a MF marker, is significantly downregulated. Our data suggest a new role of cul-4 in retinal differentiation. These studies may have significant bearings on our understanding of early eye development.


Assuntos
Olho Composto de Artrópodes/metabolismo , Proteínas Culina/metabolismo , Proteínas de Drosophila/metabolismo , Neurogênese , Animais , Olho Composto de Artrópodes/citologia , Olho Composto de Artrópodes/crescimento & desenvolvimento , Proteínas Culina/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Regulação da Expressão Gênica no Desenvolvimento , Mutação com Perda de Função , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Via de Sinalização Wnt
9.
Front Cell Dev Biol ; 8: 117, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32232042

RESUMO

Alzheimer's disease (AD, OMIM: 104300) is an age-related disorder that affects millions of people. One of the underlying causes of AD is generation of hydrophobic amyloid-beta 42 (Aß42) peptides that accumulate to form amyloid plaques. These plaques induce oxidative stress and aberrant signaling, which result in the death of neurons and other pathologies linked to neurodegeneration. We have developed a Drosophila eye model of AD by targeted misexpression of human Aß42 in the differentiating retinal neurons, where an accumulation of Aß42 triggers a characteristic neurodegenerative phenotype. In a forward deficiency screen to look for genetic modifiers, we identified a molecularly defined deficiency, which suppresses Aß42-mediated neurodegeneration. This deficiency uncovers hippo (hpo) gene, a member of evolutionarily conserved Hippo signaling pathway that regulates growth. Activation of Hippo signaling causes cell death, whereas downregulation of Hippo signaling triggers cell proliferation. We found that Hippo signaling is activated in Aß42-mediated neurodegeneration. Downregulation of Hippo signaling rescues the Aß42-mediated neurodegeneration, whereas upregulation of Hippo signaling enhances the Aß42-mediated neurodegeneration phenotypes. It is known that c-Jun-amino-terminal kinase (JNK) signaling pathway is upregulated in AD. We found that activation of JNK signaling enhances the Aß42-mediated neurodegeneration, whereas downregulation of JNK signaling rescues the Aß42-mediated neurodegeneration. We tested the nature of interactions between Hippo signaling and JNK signaling in Aß42-mediated neurodegeneration using genetic epistasis approach. Our data suggest that Hippo signaling and JNK signaling, two independent signaling pathways, act synergistically upon accumulation of Aß42 plaques to trigger cell death. Our studies demonstrate a novel role of Hippo signaling pathway in Aß42-mediated neurodegeneration.

10.
Cell Death Dis ; 7(12): e2566, 2016 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-28032862

RESUMO

In all multicellular organisms, the fundamental processes of cell proliferation and cell death are crucial for growth regulation during organogenesis. Strict regulation of cell death is important to maintain tissue homeostasis by affecting processes like regulation of cell number, and elimination of unwanted/unfit cells. The developing Drosophila eye is a versatile model to study patterning and growth, where complex signaling pathways regulate growth and cell survival. However, the molecular mechanisms underlying regulation of these processes is not fully understood. In a gain-of-function screen, we found that misexpression of cullin-4 (cul-4), an ubiquitin ligase, can rescue reduced eye mutant phenotypes. Previously, cul-4 has been shown to regulate chromatin remodeling, cell cycle and cell division. Genetic characterization of cul-4 in the developing eye revealed that loss-of-function of cul-4 exhibits a reduced eye phenotype. Analysis of twin-spots showed that in comparison with their wild-type counterparts, the cul-4 loss-of-function clones fail to survive. Here we show that cul-4 clones are eliminated by induction of cell death due to activation of caspases. Aberrant activation of signaling pathways is known to trigger cell death in the developing eye. We found that Wingless (Wg) and c-Jun-amino-terminal-(NH2)-Kinase (JNK) signaling are ectopically induced in cul-4 mutant clones, and these signals co-localize with the dying cells. Modulating levels of Wg and JNK signaling by using agonists and antagonists of these pathways demonstrated that activation of Wg and JNK signaling enhances cul-4 mutant phenotype, whereas downregulation of Wg and JNK signaling rescues the cul-4 mutant phenotypes of reduced eye. Here we present evidences to demonstrate that cul-4 is involved in restricting Wg signaling and downregulation of JNK signaling-mediated cell death during early eye development. Overall, our studies provide insights into a novel role of cul-4 in promoting cell survival in the developing Drosophila eye.


Assuntos
Proteínas Culina/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/enzimologia , Olho/citologia , Olho/enzimologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases , Animais , Caspases/metabolismo , Morte Celular , Sobrevivência Celular , Ativação Enzimática , Mutação/genética , Fenótipo , Análise de Sobrevida , Proteína Wnt1/metabolismo
11.
Nat Commun ; 7: 10972, 2016 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-26960254

RESUMO

Caspases provide vital links in non-apoptotic regulatory networks controlling inflammation, compensatory proliferation, morphology and cell migration. How caspases are activated under non-apoptotic conditions and process a selective set of substrates without killing the cell remain enigmatic. Here we find that the Drosophila unconventional myosin CRINKLED (CK) selectively interacts with the initiator caspase DRONC and regulates some of its non-apoptotic functions. Loss of CK in the arista, border cells or proneural clusters of the wing imaginal discs affects DRONC-dependent patterning. Our data indicate that CK acts as substrate adaptor, recruiting SHAGGY46/GSK3-ß to DRONC, thereby facilitating caspase-mediated cleavage and localized modulation of kinase activity. Similarly, the mammalian CK counterpart, MYO7A, binds to and impinges on CASPASE-8, revealing a new regulatory axis affecting receptor interacting protein kinase-1 (RIPK1)>CASPASE-8 signalling. Together, our results expose a conserved role for unconventional myosins in transducing caspase-dependent regulation of kinases, allowing them to take part in specific signalling events.


Assuntos
Caspase 8/metabolismo , Caspases/metabolismo , Proteínas de Drosophila/metabolismo , Miosinas/metabolismo , Animais , Linhagem Celular Tumoral , Drosophila melanogaster , Citometria de Fluxo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Imunoprecipitação , Camundongos , Microscopia Confocal , Miosina VIIa , Células NIH 3T3 , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Transdução de Sinais , Asas de Animais
12.
Curr Biol ; 26(5): 575-84, 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-26898463

RESUMO

Apoptosis-induced proliferation (AiP) is a compensatory mechanism to maintain tissue size and morphology following unexpected cell loss during normal development, and may also be a contributing factor to cancer and drug resistance. In apoptotic cells, caspase-initiated signaling cascades lead to the downstream production of mitogenic factors and the proliferation of neighboring surviving cells. In epithelial cells of Drosophila imaginal discs, the Caspase-9 ortholog Dronc drives AiP via activation of Jun N-terminal kinase (JNK); however, the specific mechanisms of JNK activation remain unknown. Here we show that caspase-induced activation of JNK during AiP depends on an inflammatory response. This is mediated by extracellular reactive oxygen species (ROSs) generated by the NADPH oxidase Duox in epithelial disc cells. Extracellular ROSs activate Drosophila macrophages (hemocytes), which in turn trigger JNK activity in epithelial cells by signaling through the tumor necrosis factor (TNF) ortholog Eiger. We propose that in an immortalized ("undead") model of AiP, signaling back and forth between epithelial disc cells and hemocytes by extracellular ROSs and TNF/Eiger drives overgrowth of the disc epithelium. These data illustrate a bidirectional cell-cell communication pathway with implication for tissue repair, regeneration, and cancer.


Assuntos
Apoptose , Proliferação de Células , Drosophila melanogaster/metabolismo , Macrófagos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Caspases/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimologia , Drosophila melanogaster/crescimento & desenvolvimento , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Larva
13.
PLoS One ; 10(9): e0137691, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26367392

RESUMO

BACKGROUND: The progressive neurodegenerative disorder Alzheimer's disease (AD) manifests as loss of cognitive functions, and finally leads to death of the affected individual. AD may result from accumulation of amyloid plaques. These amyloid plaques comprising of amyloid-beta 42 (Aß42) polypeptides results from the improper cleavage of amyloid precursor protein (APP) in the brain. The Aß42 plaques have been shown to disrupt the normal cellular processes and thereby trigger abnormal signaling which results in the death of neurons. However, the molecular-genetic mechanism(s) responsible for Aß42 mediated neurodegeneration is yet to be fully understood. METHODOLOGY/PRINCIPAL FINDINGS: We have utilized Gal4/UAS system to develop a transgenic fruit fly model for Aß42 mediated neurodegeneration. Targeted misexpression of human Aß42 in the differentiating photoreceptor neurons of the developing eye of transgenic fly triggers neurodegeneration. This progressive neurodegenerative phenotype resembles Alzheimer's like neuropathology. We identified a histone acetylase, CREB Binding Protein (CBP), as a genetic modifier of Aß42 mediated neurodegeneration. Targeted misexpression of CBP along with Aß42 in the differentiating retina can significantly rescue neurodegeneration. We found that gain-of-function of CBP rescues Aß42 mediated neurodegeneration by blocking cell death. Misexpression of Aß42 affects the targeting of axons from retina to the brain but misexpression of full length CBP along with Aß42 can restore this defect. The CBP protein has multiple domains and is known to interact with many different proteins. Our structure function analysis using truncated constructs lacking one or more domains of CBP protein, in transgenic flies revealed that Bromo, HAT and polyglutamine (BHQ) domains together are required for the neuroprotective function of CBP. This BHQ domain of CBP has not been attributed to promote survival in any other neurodegenerative disorders. CONCLUSIONS/SIGNIFICANCE: We have identified CBP as a genetic modifier of Aß42 mediated neurodegeneration. Furthermore, we have identified BHQ domain of CBP is responsible for its neuroprotective function. These studies may have significant bearing on our understanding of genetic basis of AD.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Proteína de Ligação a CREB/metabolismo , Proteínas de Drosophila/metabolismo , Retina/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/biossíntese , Peptídeos beta-Amiloides/genética , Animais , Encéfalo/patologia , Proteína de Ligação a CREB/genética , Modelos Animais de Doenças , Proteínas de Drosophila/genética , Drosophila melanogaster , Humanos , Camundongos Transgênicos , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/genética , Retina/patologia
14.
PLoS One ; 8(11): e78717, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24260128

RESUMO

Alzheimer's disease (AD, OMIM: 104300), a progressive neurodegenerative disorder with no cure to date, is caused by the generation of amyloid-beta-42 (Aß42) aggregates that trigger neuronal cell death by unknown mechanism(s). We have developed a transgenic Drosophila eye model where misexpression of human Aß42 results in AD-like neuropathology in the neural retina. We have identified an apical-basal polarity gene crumbs (crb) as a genetic modifier of Aß42-mediated-neuropathology. Misexpression of Aß42 caused upregulation of Crb expression, whereas downregulation of Crb either by RNAi or null allele approach rescued the Aß42-mediated-neurodegeneration. Co-expression of full length Crb with Aß42 increased severity of Aß42-mediated-neurodegeneration, due to three fold induction of cell death in comparison to the wild type. Higher Crb levels affect axonal targeting from the retina to the brain. The structure function analysis identified intracellular domain of Crb to be required for Aß42-mediated-neurodegeneration. We demonstrate a novel neuroprotective role of Crb in Aß42-mediated-neurodegeneration.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Axônios/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Membrana/metabolismo , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Axônios/patologia , Modelos Animais de Doenças , Proteínas de Drosophila/genética , Drosophila melanogaster , Humanos , Proteínas de Membrana/genética , Fragmentos de Peptídeos/genética , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
15.
PLoS One ; 8(11): e80829, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24282556

RESUMO

BACKGROUND: Alzheimer's disease (AD) is a debilitating age related progressive neurodegenerative disorder characterized by the loss of cognition, and eventual death of the affected individual. One of the major causes of AD is the accumulation of Amyloid-beta 42 (Aß42) polypeptides formed by the improper cleavage of amyloid precursor protein (APP) in the brain. These plaques disrupt normal cellular processes through oxidative stress and aberrant signaling resulting in the loss of synaptic activity and death of the neurons. However, the detailed genetic mechanism(s) responsible for this neurodegeneration still remain elusive. METHODOLOGY/ PRINCIPLE FINDINGS: We have generated a transgenic Drosophila eye model where high levels of human Aß42 is misexpressed in the differentiating photoreceptor neurons of the developing eye, which phenocopy Alzheimer's like neuropathology in the neural retina. We have utilized this model for a gain of function screen using members of various signaling pathways involved in the development of the fly eye to identify downstream targets or modifiers of Aß42 mediated neurodegeneration. We have identified the homeotic gene teashirt (tsh) as a suppressor of the Aß42 mediated neurodegenerative phenotype. Targeted misexpression of tsh with Aß42 in the differentiating retina can significantly rescue neurodegeneration by blocking cell death. We found that Tsh protein is absent/ downregulated in the neural retina at this stage. The structure function analysis revealed that the PLDLS domain of Tsh acts as an inhibitor of the neuroprotective function of tsh in the Drosophila eye model. Lastly, we found that the tsh paralog, tiptop (tio) can also rescue Aß42 mediated neurodegeneration. CONCLUSIONS/SIGNIFICANCE: We have identified tsh and tio as new genetic modifiers of Aß42 mediated neurodegeneration. Our studies demonstrate a novel neuroprotective function of tsh and its paralog tio in Aß42 mediated neurodegeneration. The neuroprotective function of tsh is independent of its role in retinal determination.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/toxicidade , Proteínas de Drosophila/genética , Genes Homeobox , Fragmentos de Peptídeos/toxicidade , Proteínas Repressoras/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Axônios , Morte Celular/genética , Drosophila , Microscopia Eletrônica de Varredura , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo
16.
Genesis ; 51(1): 68-74, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23109378

RESUMO

Genetic mosaic approach is commonly used in the Drosophila eye by completely abolishing or misexpressing a gene within a subset of cells to unravel its role during development. Classical genetic mosaic approach involves random clone generation in all developing fields. Consequently, a large sample size needs to be screened to generate and analyze clones in specific domains of the developing eye. To address domain specific functions of genes during axial patterning, we have developed a system for generating mosaic clones by combining Gal4/UAS and flippase (FLP)/FRT system which will allow generation of loss-of-function as well as gain-of-function clones on the dorsal and ventral eye margins. We used the bifid-Gal4 driver to drive expression of UAS-FLP. This reagent can have multiple applications in (i) studying spatio-temporal function of a gene during dorso-ventral (DV) axis specification in the eye, (ii) analyzing genetic epistasis of genes involved in DV patterning, and (iii) conducting genome wide screens in a domain specific manner.


Assuntos
Animais Geneticamente Modificados/genética , Drosophila/genética , Marcação de Genes , Mosaicismo , Animais , Olho Composto de Artrópodes/metabolismo , DNA Nucleotidiltransferases/genética , Proteínas de Drosophila/genética , Proteínas do Tecido Nervoso/genética , Proteínas com Domínio T/genética
17.
Dev Dyn ; 241(1): 69-84, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22034010

RESUMO

During organogenesis in all multi-cellular organisms, axial patterning is required to transform a single layer organ primordium into a three-dimensional organ. The Drosophila eye model serves as an excellent model to study axial patterning. Dorso-ventral (DV) axis determination is the first lineage restriction event during axial patterning of the Drosophila eye. The early Drosophila eye primordium has a default ventral fate, and the dorsal eye fate is established by onset of dorsal selector gene pannier (pnr) expression in a group of cells on the dorsal eye margin. The boundary between dorsal and ventral compartments called the equator is the site for Notch (N) activation, which triggers cell proliferation and differentiation. This review will focus on (1) chronology of events during DV axis determination; (2) how early division of eye into dorsal and ventral compartments contributes towards the growth and patterning of the fly retina, and (3) functions of DV patterning genes.


Assuntos
Padronização Corporal/fisiologia , Drosophila melanogaster/anatomia & histologia , Drosophila melanogaster/fisiologia , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Discos Imaginais/anatomia & histologia , Discos Imaginais/fisiologia , Células Fotorreceptoras de Invertebrados/citologia , Células Fotorreceptoras de Invertebrados/fisiologia
18.
Dev Biol ; 359(2): 199-208, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21920354

RESUMO

Patterning in multi-cellular organisms involves progressive restriction of cell fates by generation of boundaries to divide an organ primordium into smaller fields. We have employed the Drosophila eye model to understand the genetic circuitry responsible for defining the boundary between the eye and the head cuticle on the ventral margin. The default state of the early eye is ventral and depends on the function of Lobe (L) and the Notch ligand Serrate (Ser). We identified homothorax (hth) as a strong enhancer of the L mutant phenotype of loss of ventral eye. Hth is a MEIS class gene with a highly conserved Meis-Hth (MH) domain and a homeodomain (HD). Hth is known to bind Extradenticle (Exd) via its MH domain for its nuclear translocation. Loss-of-function of hth, a negative regulator of eye, results in ectopic ventral eye enlargements. This phenotype is complementary to the L mutant phenotype of loss-of-ventral eye. However, if L and hth interact during ventral eye development remains unknown. Here we show that (i) L acts antagonistically to hth, (ii) Hth is upregulated in the L mutant background, and (iii) MH domain of Hth is required for its genetic interaction with L, while its homeodomain is not, (iv) in L mutant background ventral eye suppression function of Hth involves novel MH domain-dependent factor(s), and (v) nuclear localization of Exd is not sufficient to mediate the Hth function in the L mutant background. Further, Exd is not a critical rate-limiting factor for the Hth function. Thus, optimum levels of L and Hth are required to define the boundary between the developing eye and head cuticle on the ventral margin.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas do Olho/metabolismo , Olho/metabolismo , Proteínas de Homeodomínio/metabolismo , Animais , Animais Geneticamente Modificados , Sítios de Ligação/genética , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Núcleo Celular/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Olho/crescimento & desenvolvimento , Proteínas do Olho/genética , Feminino , Proteínas de Homeodomínio/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteína Jagged-1 , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutação , Fenótipo , Ligação Proteica , Transporte Proteico , Proteínas Serrate-Jagged , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
19.
PLoS One ; 6(9): e24361, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21949710

RESUMO

BACKGROUND: Alzheimer's disease (AD) is an age related progressive neurodegenerative disorder. One of the reasons for Alzheimer's neuropathology is the generation of large aggregates of Aß42 that are toxic in nature and induce oxidative stress, aberrant signaling and many other cellular alterations that trigger neuronal cell death. However, the exact mechanisms leading to cell death are not clearly understood. METHODOLOGY/PRINCIPAL FINDINGS: We employed a Drosophila eye model of AD to study how Aß42 causes cell death. Misexpression of higher levels of Aß42 in the differentiating photoreceptors of fly retina rapidly induced aberrant cellular phenotypes and cell death. We found that blocking caspase-dependent cell death initially blocked cell death but did not lead to a significant rescue in the adult eye. However, blocking the levels of c-Jun NH(2)-terminal kinase (JNK) signaling pathway significantly rescued the neurodegeneration phenotype of Aß42 misexpression both in eye imaginal disc as well as the adult eye. Misexpression of Aß42 induced transcriptional upregulation of puckered (puc), a downstream target and functional read out of JNK signaling. Moreover, a three-fold increase in phospho-Jun (activated Jun) protein levels was seen in Aß42 retina as compared to the wild-type retina. When we blocked both caspases and JNK signaling simultaneously in the fly retina, the rescue of the neurodegenerative phenotype is comparable to that caused by blocking JNK signaling pathway alone. CONCLUSIONS/SIGNIFICANCE: Our data suggests that (i) accumulation of Aß42 plaques induces JNK signaling in neurons and (ii) induction of JNK contributes to Aß42 mediated cell death. Therefore, inappropriate JNK activation may indeed be relevant to the AD neuropathology, thus making JNK a key target for AD therapies.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fragmentos de Peptídeos/metabolismo , Transdução de Sinais , Animais , Morte Celular , Sobrevivência Celular , Regulação para Baixo , Drosophila melanogaster/citologia , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Discos Imaginais/crescimento & desenvolvimento , Discos Imaginais/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Fenótipo , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Retina/crescimento & desenvolvimento , Retina/metabolismo
20.
Dev Biol ; 346(2): 258-71, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20691679

RESUMO

Axial patterning is crucial for organogenesis. During Drosophila eye development, dorso-ventral (DV) axis determination is the first lineage restriction event. The eye primordium begins with a default ventral fate, on which the dorsal eye fate is established by expression of the GATA-1 transcription factor pannier (pnr). Earlier, it was suggested that loss of pnr function induces enlargement in the dorsal eye due to ectopic equator formation. Interestingly, we found that in addition to regulating DV patterning, pnr suppresses the eye fate by downregulating the core retinal determination genes eyes absent (eya), sine oculis (so) and dacshund (dac) to define the dorsal eye margin. We found that pnr acts downstream of Ey and affects the retinal determination pathway by suppressing eya. Further analysis of the "eye suppression" function of pnr revealed that this function is likely mediated through suppression of the homeotic gene teashirt (tsh) and is independent of homothorax (hth), a negative regulator of eye. Pnr expression is restricted to the peripodial membrane on the dorsal eye margin, which gives rise to head structures around the eye, and pnr is not expressed in the eye disc proper that forms the retina. Thus, pnr has dual function, during early developmental stages pnr is involved in axial patterning whereas later it promotes the head specific fate. These studies will help in understanding the developmental regulation of boundary formation of the eye field on the dorsal eye margin.


Assuntos
Padronização Corporal/genética , Proteínas de Drosophila/genética , Drosophila/embriologia , Olho/embriologia , Fatores de Transcrição/genética , Animais , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA