Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BBA Adv ; 4: 100107, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37868661

RESUMEN

C-value paradox refers to the lack of correlation between biological complexity and the intuitively expected protein-coding genomic information or DNA content. Here I discuss five questions about this paradox: i) Do biologically complex organisms carry more protein-coding genes? ii) Does variable accumulation of selfish/ junk/ parasitic DNA underlie the c-value paradox? iii) Can nucleoskeletal or nucleotypic function of DNA explain the enigma of orders of magnitude high levels of DNA in some 'lower' taxa or in taxonomically related species? iv) Can the newly understood noncoding but functional DNA explain the c-value paradox? and, v) Does natural selection uniformly apply the anthropocentric parameters for 'optimum' and 'economy'? Answers to Q.1-5 are largely negative. Biology presents numerous 'anomalous' examples where the same end function/ phenotype is attained in different organisms through astoundingly diverse ways that appear 'illogical' in our perceptions. Such evolutionary oddities exist because natural selection, unlike a designer, exploits random and stochastic events to modulate the existing system. Consequently, persistence of the new-found 'solution/s' often appear bizarre, uneconomic, and therefore, paradoxical to human logic. The unexpectedly high c-values in diverse organisms are irreversible evolutionary accidents that persisted, and the additional DNA often got repurposed over the evolutionary time scale. Therefore, the c-value paradox is a redundant issue. Future integrative biological studies should address evolutionary mechanisms and processes underlying sporadic DNA expansions/ contractions, and how the newly acquired DNA content has been repurposed in diverse groups.

2.
Indian J Med Ethics ; VIII(2): 143-145, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36880461

RESUMEN

Kishor Patwardhan's 'confession' in this journal [1] has initiated the expected debate, which I hope leads to some good developments for the teaching and practice of Ayurveda. Before, commenting on this issue, I should myself confess that I am neither formally trained in Ayurveda nor practising it. A basic research interest in Ayurvedic biology [2] led me to learn about the "fundamental principles" of Ayurveda and to experimentally examine effects of some Ayurvedic formulations using animal models like Drosophila and mouse at organismic, cellular, and molecular levels. During the past 16 to 17 years of my active engagement with Ayurvedic Biology, I had multiple opportunities to discuss the principles and philosophies of Ayurveda with formally trained Ayurvedacharyas and others who have an interest in this classical healthcare system. These experiences enhanced my appreciation of the wisdom of ancient scholars that led them to methodically compile the elaborate details of treatment for various health conditions in the classical Samhitas and, as noted earlier [3], gave me a "ring-side" view of Ayurveda. Despite the above limitations, an advantage of the "ring-side" view is the possibility of comprehending the philosophies and practices prevalent in Ayurveda in an unbiased manner and weighing them against contemporary practices in other disciplines.


Asunto(s)
Medicina Ayurvédica , Humanos , Ratones , Animales
3.
Methods Mol Biol ; 2609: 271-293, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36515841

RESUMEN

Heterogeneous nuclear ribonucleoproteins (hnRNPs) are a family of RNA-binding proteins that modulate multiple aspects of gene activity and RNA processing, including transcription, splicing, localization, translation, and decay of RNA. Interaction of hnRNPs with RNA is a highly dynamic but regulated process. Poly(ADP-ribose) polymerase (PARP)-dependent PARylation of different hnRNPs is a well-known posttranslational modification that affects their interactions with RNA. Here, we described a protocol for in situ localization of RNA-binding proteins (RBPs) on giant polytene chromosomes in Drosophila larval salivary glands, which have been widely used to visualize the dynamic binding profiles of various RBPs and other transcription-related proteins at specific loci on chromosomes. This chapter also includes a stepwise description of RNA:RNA in situ hybridization, in conjunction with immunostaining, using polytene chromosome squashes or intact tissues. We also highlight advanced live cell imaging methods, including FRAP and FLIP, using transgenic lines that express fluorescent-tagged hnRNPs. These cytological approaches can be used to visualize the localization of RNA-binding proteins and their interacting RNAs under different cellular conditions.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Cromosomas Politénicos/genética , Cromosomas Politénicos/metabolismo , ARN/metabolismo
4.
J Genet ; 1012022.
Artículo en Inglés | MEDLINE | ID: mdl-36330787

RESUMEN

The pioneering studies carried out on heat shock-induced synthesis of specific proteins in the early 1970s did not identify any Hsp60 family protein in Drosophila. By the early 1980s, although the members of Hsp60 family of heat shock proteins (Hsp) were identified in a wide range of eukaryotes as homologs of the bacterial GroEL, none was known in Drosophila. The existence of the Hsp60 family protein was serendipitously revealed in Drosophila in my laboratory in 1989. Contrary to the earlier reports that all tissues in flies display the canonical heat shock response, the larval Malpighian tubules (MT) did not show induction of any of the major Hsps but synthesis of a putative Hsp60 family protein was found to be the most abundant in this tissue. A few years later, we identified this MTspecific heat shock-induced protein to indeed be a member of the Hsp60/chaperonin family. The Drosophila genome sequence projects subsequently revealed four putative Hsp60 gene sequences in the D. melanogaster genome. The present historical perspective chronicles contributions from my and other laboratories that unraveled several aspects of intriguing biology of the multiple Hsp60 genes in D. melanogaster, and highlights challenging questions awaiting future studies.


Asunto(s)
Chaperonina 60 , Drosophila melanogaster , Animales , Chaperonina 60/genética , Chaperonina 60/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Drosophila/genética , Proteínas de Choque Térmico/genética , Respuesta al Choque Térmico , Proteínas HSP70 de Choque Térmico/genética
5.
Cell Stress Chaperones ; 27(4): 431-448, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35704239

RESUMEN

Cancer cells generally overexpress heat shock proteins (Hsps), the major components of cellular stress response, to overcome and survive the diverse stresses. However, the specific roles of Hsps in initiation and establishment of cancers remain unclear. Using loss of Lgl-mediated epithelial tumorigenesis in Drosophila, we induced tumorigenic somatic clones of different genetic backgrounds to examine the temporal and spatial expression and roles of major heat shock proteins in tumor growth. The constitutively expressed Hsp83, Hsc70 (heat shock cognate), Hsp60 and Hsp27 show elevated levels in all cells of the tumorigenic clone since early stages, which persists till their transformation. However, the stress-inducible Hsp70 is expressd only in a few cells at later stage of established tumorous clones that show high F-actin aggregation. Intriguingly, levels of heat shock factor (HSF), the master regulator of Hsps, remain unaltered in these tumorous cells and its down-regulation does not affect tumorigenic growth of lgl- clones overexpressing Yorkie, although down-regulation of Hsp83 prevents their survival and growth. Interestingly, overexpression of HSF or Hsp83 in lgl- cells makes them competitively successful in establishing tumorous clones. These results show that the major constitutively expressed Hsps, but not the stress-inducible Hsp70, are involved in early as well as late stages of epithelial tumors and their elevated expression in lgl- clones co-overexpressing Yorkie is independent of HSF.


Asunto(s)
Drosophila , Neoplasias , Animales , Proteínas de Choque Térmico HSP27 , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas de Choque Térmico/genética , Respuesta al Choque Térmico/genética , Neoplasias/genética
6.
J Genet ; 1002021.
Artículo en Inglés | MEDLINE | ID: mdl-34622796

RESUMEN

Early genetic studies with Drosophila revealed similar mutant phenotypes for many X-linked genes, in males with one and in females with two copies of the mutant allele following the XY/XX mode of sex determination. These observations led to evocation of the phenomenon of dosage compensation. By the 1960s, contrasting theories were advanced by H. J. Muller and R. B. Goldschmidt to explain the equalized expression of many X-linked genes despite their dosage difference in male and female flies. Evidence from genetic studies led Muller to propose existence of many modifiers whose action on individual X-linked genes resulted, through a 'piecemeal' regulation, in equalized expression of the dosage compensated X-linked genes, while Goldschmidt believed that invocation of multiple modifiers or compensators was unnecessary since dosage compensation was a direct outcome of the sex-specific physiologies of male and female flies. Muller did not agree with some cytological studies that suggested that the single X-chromosome in male cells works twice as hard as each of the two X-chromosomes in female cells (hyperactive male X model), but preferred partial repression of each X-chromosome in female flies. This historical perspective relates these divergent theories with my own doctoral work in A. S. Mukherjee's laboratory at Calcutta University, which, while ruling out Golschmidt's sex-physiology theory, established cell-autonomous regulation of the earlier proposed hyperactivity of the single X in male Drosophila in a piecemeal manner.


Asunto(s)
Cromosomas de Insectos , Compensación de Dosificación (Genética) , Drosophila/genética , Genética/historia , Animales , Femenino , Genes Ligados a X , Historia del Siglo XX , India , Masculino , Modelos Genéticos , Cromosomas Politénicos , Cromosoma X
7.
Dev Dyn ; 248(12): 1211-1231, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31415125

RESUMEN

BACKGROUND: Dilp8-mediated inhibition of ecdysone synthesis and pupation in holometabolous insects maintains developmental homeostasis through stringent control of timing and strength of molting signals. We examined reasons for normal pupation but early pupal death observed in certain cases. RESULTS: Overexpression of activated Ras in developing eye/wing discs inhibited Ptth expression in brain via upregulated JNK signaling mediated Dilp8 secretion from imaginal discs, which inhibited ecdysone synthesis in prothoracic gland after pupariation, leading to death of ~25- to 30-hour-old pupae. Inhibition of elevated Ras signaling completely rescued early pupal death while post-pupation administration of ecdysone to organisms with elevated Ras signaling in eye discs partially rescued their early pupal death. Unlike the earlier known Dilp8 action in delaying pupation, hyperactivated Ras mediated elevation of pJNK signaling in imaginal discs caused Dilp8 secretion after pupariation. Ectopic expression of certain other transgene causing pupal lethality similarly enhanced pJNK and early pupal Dilp8 levels. Suboptimal ecdysone levels after 8 hours of pupation prevented the early pupal metamorphic changes and caused organismal death. CONCLUSIONS: Our results reveal early pupal stage as a novel Dilp8 mediated post-pupariation checkpoint and provide further evidence for interorgan signaling during development, wherein a peripheral tissue influences the CNS driven endocrine function.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster , IMP Deshidrogenasa/metabolismo , Discos Imaginales/embriología , Discos Imaginales/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Homeostasis/genética , IMP Deshidrogenasa/genética , Discos Imaginales/crecimiento & desarrollo , Péptidos y Proteínas de Señalización Intercelular/genética , Larva , Sistema de Señalización de MAP Quinasas/fisiología , Organogénesis/genética , Pupa/genética , Pupa/crecimiento & desarrollo , Transducción de Señal/genética , Proteínas Activadoras de ras GTPasa/metabolismo
8.
J Biosci ; 44(2)2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31180049

RESUMEN

We examined interactions between the 83 kDa heat-shock protein (Hsp83) and hsrω long noncoding RNAs (lncRNAs) in hsrω66 Hsp90GFP homozygotes, which almost completely lack hsrω lncRNAs but over-express Hsp83. All +/+; hsrω66 Hsp90GFP progeny died before the third instar. Rare Sp/CyO; hsrω66 Hsp90GFP reached the third instar stage but phenocopied l(2)gl mutants, becoming progressively bulbous and transparent with enlarged brain and died after prolonged larval life. Additionally, ventral ganglia too were elongated. However, hsrω66 Hsp90GFP/TM6B heterozygotes, carrying +/+ or Sp/CyO second chromosomes, developed normally. Total RNA sequencing (+/+, +/+; hsrω66/hsrω66, Sp/CyO; hsrω66/ hsrω66, +/+; Hsp90GFP/Hsp90GFP and Sp/CyO; hsrω66 Hsp90GFP/hsrω66 Hsp90GFP late third instar larvae) revealed similar effects on many genes in hsrω66 and Hsp90GFP homozygotes. Besides additive effect on many of them, numerous additional genes were affected in Sp/CyO; hsrω66 Hsp90GFP larvae, with l(2)gl and several genes regulating the central nervous system being highly down-regulated in surviving Sp/CyO; hsrω66 Hsp90GFP larvae, but not in hsrω66 or Hsp90GFP single mutants. Hsp83 and several omega speckle-associated hnRNPs were bioinformatically found to potentially bind with these gene promoters and transcripts. Since Hsp83 and hnRNPs are also known to interact, elevated Hsp83 in an altered background of hnRNP distribution and dynamics, due to near absence of hsrω lncRNAs and omega speckles, can severely perturb regulatory circuits with unexpected consequences, including down-regulation of tumoursuppressor genes such as l(2)gl.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Choque Térmico/genética , Larva/genética , ARN Largo no Codificante/genética , Mutaciones Letales Sintéticas , Animales , Animales Modificados Genéticamente , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Ganglios de Invertebrados/crecimiento & desarrollo , Ganglios de Invertebrados/metabolismo , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Choque Térmico/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Heterocigoto , Homocigoto , Larva/crecimiento & desarrollo , Larva/metabolismo , Fenotipo , ARN Largo no Codificante/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transcriptoma
9.
Gene Expr Patterns ; 33: 20-36, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31096022

RESUMEN

We exploited the high Ras activity induced differentiation of supernumerary R7 cells in Drosophila eyes to examine if hsrω lncRNAs influence active Ras signaling. Surprisingly, either down- or up-regulation of hsrω lncRNAs in sev-GAL4>RasV12 expressing eye discs resulted in complete pupal lethality and substantially greater increase in R7 photoreceptor number at the expense of cone cells. Enhanced nuclear p-MAPK and presence of sev-GAL4 driven RasV12 bound RafRBDFLAG in cells not expressing the sev-GAL4 driver indicated non-cell autonomous spread of Ras signaling when hsrω levels were co-altered. RNA-sequencing revealed that down-and up-regulation of hsrω transcripts in sev-GAL4>RasV12 expressing eye discs elevated transcripts of positive or negative modulators, respectively, of Ras signaling so that either condition enhances it. Altered hsrω transcript levels in sev-GAL4>RasV12 expressing discs also affected sn/sno/sca RNAs and some other RNA processing transcript levels. Post-transcriptional changes due to the disrupted intra-cellular dynamicity of omega speckle associated hnRNPs and other RNA-binding proteins that follow down- or up-regulation of hsrω lncRNAs appear to be responsible for the further elevated Ras signaling. Cell autonomous and non-autonomous enhancement of Ras signaling by lncRNAs like hsrω has implications for cell signaling during high Ras activity commonly associated with some cancers.


Asunto(s)
Proteínas de Drosophila/genética , Células Fotorreceptoras de Invertebrados/metabolismo , ARN Largo no Codificante/genética , Proteínas ras/genética , Animales , Diferenciación Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Sistema de Señalización de MAP Quinasas , Células Fotorreceptoras de Invertebrados/citología , ARN Largo no Codificante/metabolismo , Proteínas ras/metabolismo
10.
Front Mol Neurosci ; 10: 323, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29089867

RESUMEN

Alzheimer's disease (AD) is an age-related neurodegenerative disorder, characterized by progressive loss of cognitive functions and memory. Excessive intake of aluminum chloride in drinking water is associated with amyloid plaques and neurofibrillary tangles in the brain, which are the hallmark of AD. We have evaluated brain energy metabolism in aluminum chloride (AlCl3) mouse model of AD. In addition, effectiveness of Rasa Sindoor (RS), a formulation used in Indian Ayurvedic medicine, for alleviation of symptoms of AD was evaluated. Mice were administered AlCl3 (40 mg/kg) intraperitoneally once a day for 60 days. The memory of mice was measured using Morris Water Maze test. The 13C labeling of brain amino acids was measured ex vivo in tissue extracts using 1H-[13C]-NMR spectroscopy with timed infusion of [1,6-13C2]glucose. The 13C turnover of brain amino acids was analyzed using a three-compartment metabolic model to derive the neurotransmitter cycling and TCA cycle rates associated with glutamatergic and GABAergic pathways. Exposure of AlCl3 led to reduction in memory of mice. The glutamatergic and GABAergic neurotransmitter cycling and glucose oxidation were found to be reduced in the cerebral cortex, hippocampus, and striatum following chronic AlCl3 treatment. The perturbation in metabolic rates was highest in the cerebral cortex. However, reduction in metabolic fluxes was higher in hippocampus and striatum following one month post AlCl3 treatment. Most interestingly, oral administration of RS (2 g/kg) restored memory as well as the energetics of neurotransmission in mice exposed to AlCl3. These data suggest therapeutic potential of RS to manage cognitive functions and memory in preclinical AD.

11.
Adv Exp Med Biol ; 1008: 75-118, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28815537

RESUMEN

Recent years have witnessed a remarkable interest in exploring the significance of pervasive noncoding transcripts in diverse eukaryotes. Classical cytogenetic studies using the Drosophila model system unraveled the perplexing attributes and "functions" of the "gene"-poor heterochromatin. Recent molecular studies in the fly model are likewise revealing the very diverse and significant roles played by long noncoding RNAs (lncRNAs) in development, gene regulation, chromatin organization, cell and nuclear architecture, etc. There has been a rapid increase in the number of identified lncRNAs, although a much larger number still remains unknown. The diversity of modes of actions and functions of the limited number of Drosophila lncRNAs, which have been examined, already reflects the profound roles of such RNAs in generating and sustaining the biological complexities of eukaryotes. Several of the known Drosophila lncRNAs originate as independent sense or antisense transcripts from promoter or intergenic, intronic, or 5'/3'-UTR regions, while many of them are independent genes that produce only lncRNAs or coding as well as noncoding RNAs. The different lncRNAs affect chromatin organization (local or large-scale pan-chromosomal), transcription, RNA processing/stability, or translation either directly through interaction with their target DNA sequences or indirectly by acting as intermediary molecules for specific regulatory proteins or may act as decoys/sinks, or storage sites for specific proteins or groups of proteins, or may provide a structural framework for the assembly of substructures in nucleus/cytoplasm. It is interesting that many of the "functions" alluded to heterochromatin in earlier cytogenetic studies appear to find correlates with the known subtle as well as far-reaching actions of the different small and long noncoding RNAs. Further studies exploiting the very rich and powerful genetic and molecular resources available for the Drosophila model are expected to unravel the mystery underlying the long reach of ncRNAs.


Asunto(s)
Heterocromatina , Biosíntesis de Proteínas/fisiología , Procesamiento Postranscripcional del ARN/fisiología , ARN Largo no Codificante , Transcripción Genética/fisiología , Animales , Drosophila melanogaster , Heterocromatina/genética , Heterocromatina/metabolismo , ARN Largo no Codificante/sangre , ARN Largo no Codificante/clasificación , ARN Largo no Codificante/genética
12.
J Biosci ; 42(3): 363-371, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29358550

RESUMEN

Alzheimer's disease (AD) is the most common neurodegenerative disorder characterized by progressive loss of memory and cognitive function. The cerebral metabolic rate of glucose oxidation has been shown to be reduced in AD. The present study evaluated efficacy of dietary Amalaki Rasayana (AR), an Ayurvedic formulation used in Indian traditional system, in AbPP-PS1 mouse model of AD in ameliorating memory and neurometabolism, and compared with donepezil, a standard FDA approved drug for AD. The memory of mice was measured using Morris Water Maze analysis. The cerebral metabolism was followed by 13C labelling of brain amino acids in tissue extracts ex vivo using 1H-[13C]-NMR spectroscopy together with a short time infusion of [1,6-13C2]glucose to mice. The intervention with Amalaki Rasayana showed improved learning and memory in AbPP-PS1 mice. The 13C labelings of GluC4, GABAC2 and GlnC4 were reduced in AbPP-PS1 mice when compared with wild-type controls. Intervention of AR increased the 13C labelling of amino acids suggesting a significant enhancement in glutamatergic and GABAergic metabolic activity in AbPP-PS1 mice similar to that observed with donepezil treatment. These data suggest that AR has potential to improve memory and cognitive function in AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Cognición/efectos de los fármacos , Memoria/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Extractos Vegetales/farmacología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Isótopos de Carbono , Donepezilo , Expresión Génica , Glucosa/metabolismo , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Humanos , Indanos/farmacología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Medicina Ayurvédica/métodos , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Piperidinas/farmacología , Presenilina-1/genética , Presenilina-1/metabolismo , Ácido gamma-Aminobutírico/metabolismo
13.
J Biosci ; 41(4): 697-711, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27966490

RESUMEN

Amalaki Rasayana (AR) is a common Ayurvedic herbal formulation of Phyllanthus emblica fruits and some other ingredients, and is used for general good health and healthy aging. We reported it to improve life history traits and to suppress neurodegeneration as well as induced apoptosis in Drosophila. The present study examines responses of Drosophila reared on AR-supplemented food to crowding, thermal or oxidative stresses. Wild-type larvae/flies reared on AR-supplemented food survived the various cell stresses much better than those reared on control food. AR-fed mutant park13 or DJ-1 beta Delta93 (Parkinson's disease model) larvae/flies, however, showed only partial or no protection, respectively, against paraquat-induced oxidative stress, indicating essentiality of DJ-1 beta for AR-mediated oxidative stress tolerance. AR feeding reduced the accumulation of reactive oxygen species (ROS) and lipid peroxidation even in aged (35-day-old) wild-type flies while enhancing superoxide dismutase (SOD) activity. We show that while Hsp70 or Hsp83 expression under normal or stress conditions was not affected by AR feeding, Hsp27 levels were elevated in AR-fed wild-type control as well as heat-shocked larvae. Therefore, besides the known anti-oxidant activity of Phyllanthus emblica fruits, dietary AR also enhances cellular levels of Hsp27. Our in vivo study on a model organism shows that AR feeding significantly improves tolerance to a variety of cell stresses through reduced ROS and lipid peroxidation on the one hand, and enhanced SOD activity and Hsp27 on the other. The resulting better homeostasis improves life span and quality of organism's life.


Asunto(s)
Proteínas de Drosophila/biosíntesis , Proteínas de Choque Térmico HSP27/biosíntesis , Proteínas HSP70 de Choque Térmico/biosíntesis , Proteínas de Choque Térmico/biosíntesis , Enfermedad de Parkinson/tratamiento farmacológico , Extractos Vegetales/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteínas de Choque Térmico HSP27/genética , Proteínas HSP70 de Choque Térmico/genética , Proteínas de Choque Térmico/genética , Humanos , Larva/efectos de los fármacos , Larva/genética , Larva/crecimiento & desarrollo , Medicina Ayurvédica , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/biosíntesis
14.
Oncotarget ; 7(48): 78281-78296, 2016 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-27835876

RESUMEN

Aggregation of proteins with the expansion of polyglutamine tracts in the brain underlies progressive genetic neurodegenerative diseases (NDs) like Huntington's disease and spinocerebellar ataxias (SCA). An insensitive cellular proteotoxic stress response to non-native protein oligomers is common in such conditions. Indeed, upregulation of heat shock factor 1 (HSF1) function and its target protein chaperone expression has shown promising results in animal models of NDs. Using an HSF1 sensitive cell based reporter screening, we have isolated azadiradione (AZD) from the methanolic extract of seeds of Azadirachta indica, a plant known for its multifarious medicinal properties. We show that AZD ameliorates toxicity due to protein aggregation in cell and fly models of polyglutamine expansion diseases to a great extent. All these effects are correlated with activation of HSF1 function and expression of its target protein chaperone genes. Notably, HSF1 activation by AZD is independent of cellular HSP90 or proteasome function. Furthermore, we show that AZD directly interacts with purified human HSF1 with high specificity, and facilitates binding of HSF1 to its recognition sequence with higher affinity. These unique findings qualify AZD as an ideal lead molecule for consideration for drug development against NDs that affect millions worldwide.


Asunto(s)
ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Factores de Transcripción del Choque Térmico/metabolismo , Limoninas/farmacología , Enfermedades Neurodegenerativas/prevención & control , Fármacos Neuroprotectores/farmacología , Péptidos/metabolismo , Extractos Vegetales/farmacología , Agregación Patológica de Proteínas , Animales , Azadirachta/química , ADN/genética , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Células HCT116 , Células HEK293 , Factores de Transcripción del Choque Térmico/genética , Humanos , Limoninas/aislamiento & purificación , Limoninas/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Fármacos Neuroprotectores/aislamiento & purificación , Fármacos Neuroprotectores/metabolismo , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/metabolismo , Unión Proteica , Semillas , Factores de Tiempo , Transfección
15.
Cell Stress Chaperones ; 21(1): 105-120, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26386576

RESUMEN

A delayed organismic lethality was reported in Drosophila following heat shock when developmentally active and stress-inducible noncoding hsrω-n transcripts were down-regulated during heat shock through hs-GAL4-driven expression of the hsrω-RNAi transgene, despite the characteristic elevation of all heat shock proteins (Hsp), including Hsp70. Here, we show that hsrω-RNAi transgene expression prior to heat shock singularly prevents accumulation of Hsp70 in all larval tissues without affecting transcriptional induction of hsp70 genes and stability of their transcripts. Absence of the stress-induced Hsp70 accumulation was not due to higher levels of Hsc70 in hsrω-RNAi transgene-expressing tissues. Inhibition of proteasomal activity during heat shock restored high levels of the induced Hsp70, suggesting very rapid degradation of the Hsp70 even during the stress when hsrω-RNAi transgene was expressed ahead of heat shock. Unexpectedly, while complete absence of hsrω transcripts in hsrω (66) homozygotes (hsrω-null) did not prevent high accumulation of heat shock-induced Hsp70, hsrω-RNAi transgene expression in hsrω-null background blocked Hsp70 accumulation. Nonspecific RNAi transgene expression did not affect Hsp70 induction. These observations reveal that, under certain conditions, the stress-induced Hsp70 can be selectively and rapidly targeted for proteasomal degradation even during heat shock. In the present case, the selective degradation of Hsp70 does not appear to be due to down-regulation of the hsrω-n transcripts per se; rather, this may be an indirect effect of the expression of hsrω-RNAi transgene whose RNA products may titrate away some RNA-binding proteins which may also be essential for stability of the induced Hsp70.


Asunto(s)
Drosophila melanogaster/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Respuesta al Choque Térmico/genética , ARN Largo no Codificante/genética , Animales , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas HSP70 de Choque Térmico/genética , Larva/genética , Larva/metabolismo , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Activación Transcripcional/genética
16.
Chromosoma ; 125(3): 373-88, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26373285

RESUMEN

Unlike the telomerase-dependent mammalian telomeres, HeT-A, TART, and TAHRE (HTT) retroposon arrays regulate Drosophila telomere length. Cap prevents telomeric associations (TAs) and telomeric fusions (TFs). Our results suggest important roles of Hrb87F in telomeric HTT array and cap maintenance in Drosophila. All chromosome arms, except 2L, in Df(3R)Hrb87F homozygotes (Hrb87F-null) displayed significantly elongated telomeres with amplified HTT arrays and high TAs, all of which resolved without damage. Presence of FLAG-tagged Hrb87F (FLAG-Hrb87F) on cap and subtelomeric regions following hsFLAG-Hrb87F transgene expression in Df(3R)Hrb87F homozygotes suppressed TAs without affecting telomere length. A normal X-chromosome telomere expanded within five generations in Hrb87F-null background and displayed high TAs, but not when hsFLAG-Hrb87F was co-expressed. Tel (1) /Gaiano line or HP1 loss-of-function mutant-derived expanded telomeres carry Hrb87F on cap and HTT arrays while Hrb87F-null telomeres have HP1 and HOAP on caps and expanded HTT arrays. ISWI, seen only on cap on normal telomeres, was abundant on Hrb87F-null expanded HTT arrays. Extended telomeres derived from Tel (1) (Gaiano) or HP1-null mutation background interact with those from Hrb87F-null, since while the end association frequency was negligible in Df(3R)Hrb87F/+ nuclei, it increased significantly in co-presence of Tel (1) or HP1-null-based expanded telomere/s. Together, these suggest complex interactions between members of the proteome of telomere so that absence of any key member leads to telomere expansion and/or enhanced TAs/TFs. HTT expansion in Hrb87F-null condition is not developmental but a germline event presumably because absence of Hrb87F in germline may deregulate HTT retroposition/replication leading to telomere elongation.


Asunto(s)
Proteínas de Drosophila/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Proteínas Nucleares/metabolismo , Homeostasis del Telómero/fisiología , Telómero/metabolismo , Cromosoma X/metabolismo , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster , Ribonucleoproteínas Nucleares Heterogéneas/genética , Proteínas Nucleares/genética , Telómero/genética , Cromosoma X/genética
18.
J Genet ; 94(4): 575-84, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26690511

RESUMEN

Organisms with heterochromatic sex chromosomes need to compensate for differences in dosages of the sex chromosome-linked genes that have somatic functions. In-depth cytological and subsequent biochemical and molecular studies on dosage compensation started with Mary F. Lyon's proposal in early 1960s that the Barr body in female mammalian somatic cells represented one of the randomly inactivated and heterochromatinized X chromosomes. In contrast,Drosophila was soon shown to achieve dosage compensation through hypertranscription of single X in male whose chromatin remains more open.Identification of proteins that remodel chromatin either to cause one of the two X chromosomes in somatic cells of very early female mammalian embryos to become condensed and inactive or to remodel the single X in male Drosophila embryos to a more open state for hypertranscription provided important insights into the underlying cellular epigenetic processes.However, the most strikin g and unexpected discoveries were the identification of long noncoding RNAs (lncRNAs), X-inactive specific transcript (Xist) in mammals and roX1/2 in Drosophila, which were essential for achieving the contrasting chromatin organizations but leading to similar end result in terms of dosage compensation of X-linked genes in females and males. An overview of the processes of X inactivation or hyperactivation in mammals and Drosophila, respectively, and the roles played by Xist, roX1/2 and other lncRNAs in these events is presented.


Asunto(s)
Compensación de Dosificación (Genética)/genética , Drosophila/genética , Mamíferos/genética , ARN Largo no Codificante/genética , Cromosoma X/genética , Animales , Epigénesis Genética/genética , Humanos
19.
J Genet ; 94(3): 407-16, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26440079

RESUMEN

The binary GAL4-UAS system of conditional gene expression is widely used by Drosophila geneticists to target expression of the desired transgene in tissue of interest. In many studies, a preferred target tissue is the Drosophila eye, for which the sev-GAL4 and GMR-GAL4 drivers are most widely used since they are believed to be expressed exclusively in the developing eye cells. However, several reports have noted lethality following expression of certain transgenes under these GAL4 drivers notwithstanding the fact that eye is not essential for survival of the fly. Therefore, to explore the possibility that these drivers may also be active in tissues other than eye, we examined the expression of UAS-GFP reporter driven by the sev-GAL4 or GMR-GAL4 drivers. We found that both these drivers are indeed expressed in additional tissues, including a common set of specific neuronal cells in larval and pupal ventral and cerebral ganglia. Neither sev nor glass gene has so far been reported to be expressed in these neuronal cells. Expression pattern of sev-GAL4 driver parallels that of the endogenous Sevenless protein. In addition to cells in which sev-GAL4 is expressed, the GMR-GAL4 is expressed in several other larval cell types also. Further, two different GMR-GAL4 lines also show some specific differences in their expression domains outside the eye discs. These findings emphasize the need for a careful confirmation of the expression domains of a GAL4 driver being used in a given study, rather than relying only on the empirically claimed expression domains.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción/genética , Animales , Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/embriología , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Ganglios de Invertebrados/citología , Ganglios de Invertebrados/metabolismo , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Larva/citología , Larva/metabolismo , Neuronas/citología , Neuronas/metabolismo , Especificidad de Órganos/genética , Pupa/citología , Pupa/metabolismo , Factores de Transcripción/metabolismo
20.
Ann Neurosci ; 22(2): 55-7, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26130907
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...