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.
Am J Physiol Heart Circ Physiol ; 326(2): H433-H440, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38099848

RESUMO

Pulmonary and systemic congestion as a consequence of heart failure are clinically recognized as alarm signals for clinical outcome and mortality. Although signs and symptoms of congestion are well detectable in patients, monitoring of congestion in small animals with heart failure lacks adequate noninvasive methodology yet. Here, we developed a novel ultrasonography-based scoring system to assess pulmonary and systemic congestion in experimental heart failure, by using lung ultrasound (LUS) and imaging of the inferior vena cava (Cava), termed CavaLUS. CavaLUS was established and tested in a rat model of supracoronary aortic banding and a mouse model of myocardial infarction, providing high sensitivity and specificity while correlating to numerous parameters of cardiac performance and disease severity. CavaLUS, therefore, provides a novel comprehensive tool for experimental heart failure in small animals to noninvasively assess congestion.NEW & NOTEWORTHY As thorough, noninvasive assessment of congestion is not available in small animals, we developed and validated an ultrasonography-based research tool to evaluate pulmonary and central venous congestion in experimental heart failure models.


Assuntos
Insuficiência Cardíaca , Hiperemia , Humanos , Camundongos , Animais , Ratos , Hiperemia/diagnóstico por imagem , Pulmão/diagnóstico por imagem , Ultrassonografia/métodos , Insuficiência Cardíaca/diagnóstico por imagem , Insuficiência Cardíaca/etiologia , Veia Cava Inferior/diagnóstico por imagem
2.
Nat Commun ; 14(1): 4416, 2023 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-37479718

RESUMO

Pulmonary hypertension worsens outcome in left heart disease. Stiffening of the pulmonary artery may drive this pathology by increasing right ventricular dysfunction and lung vascular remodeling. Here we show increased stiffness of pulmonary arteries from patients with left heart disease that correlates with impaired pulmonary hemodynamics. Extracellular matrix remodeling in the pulmonary arterial wall, manifested by dysregulated genes implicated in elastin degradation, precedes the onset of pulmonary hypertension. The resulting degradation of elastic fibers is paralleled by an accumulation of fibrillar collagens. Pentagalloyl glucose preserves arterial elastic fibers from elastolysis, reduces inflammation and collagen accumulation, improves pulmonary artery biomechanics, and normalizes right ventricular and pulmonary hemodynamics in a rat model of pulmonary hypertension due to left heart disease. Thus, targeting extracellular matrix remodeling may present a therapeutic approach for pulmonary hypertension due to left heart disease.


Assuntos
Cardiopatias , Hipertensão Pulmonar , Humanos , Animais , Ratos , Artéria Pulmonar , Fenômenos Biomecânicos , Elastina
3.
Front Immunol ; 13: 959209, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36275740

RESUMO

Pulmonary hypertension (PH) is a progressive disease that arises from multiple etiologies and ultimately leads to right heart failure as the predominant cause of morbidity and mortality. In patients, distinct inflammatory responses are a prominent feature in different types of PH, and various immunomodulatory interventions have been shown to modulate disease development and progression in animal models. Specifically, PH-associated inflammation comprises infiltration of both innate and adaptive immune cells into the vascular wall of the pulmonary vasculature-specifically in pulmonary vascular lesions-as well as increased levels of cytokines and chemokines in circulating blood and in the perivascular tissue of pulmonary arteries (PAs). Previous studies suggest that altered hemodynamic forces cause lung endothelial dysfunction and, in turn, adherence of immune cells and release of inflammatory mediators, while the resulting perivascular inflammation, in turn, promotes vascular remodeling and the progression of PH. As such, a vicious cycle of endothelial activation, inflammation, and vascular remodeling may develop and drive the disease process. PA stiffening constitutes an emerging research area in PH, with relevance in PH diagnostics, prognostics, and as a therapeutic target. With respect to its prognostic value, PA stiffness rivals the well-established measurement of pulmonary vascular resistance as a predictor of disease outcome. Vascular remodeling of the arterial extracellular matrix (ECM) as well as vascular calcification, smooth muscle cell stiffening, vascular wall thickening, and tissue fibrosis contribute to PA stiffening. While associations between inflammation and vascular stiffening are well-established in systemic vascular diseases such as atherosclerosis or the vascular manifestations of systemic sclerosis, a similar connection between inflammatory processes and PA stiffening has so far not been addressed in the context of PH. In this review, we discuss potential links between inflammation and PA stiffening with a specific focus on vascular calcification and ECM remodeling in PH.


Assuntos
Hipertensão Pulmonar , Calcificação Vascular , Doenças Vasculares , Animais , Hipertensão Pulmonar/etiologia , Artéria Pulmonar , Remodelação Vascular , Inflamação , Citocinas , Mediadores da Inflamação
4.
J Vis Exp ; (181)2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35311828

RESUMO

Pulmonary hypertension due to left heart disease (PH-LHD) is the most common form of PH, yet its pathophysiology is poorly characterized than pulmonary arterial hypertension (PAH). As a result, approved therapeutic interventions for the treatment or prevention of PH-LHD are missing. Medications used to treat PH in PAH patients are not recommended for treatment of PH-LHD, as reduced pulmonary vascular resistance (PVR) and increased pulmonary blood flow in the presence of increased left-sided filling pressures may cause left heart decompensation and pulmonary edema. New strategies need to be developed to reverse PH in LHD patients. In contrast to PAH, PH-LHD develops due to increased mechanical load caused by congestion of blood into the lung circulation during left heart failure. Clinically, mechanical unloading of the left ventricle (LV) by aortic valve replacement in aortic stenosis patients or by implantation of LV assist devices in end-stage heart failure patients normalizes not only pulmonary arterial and right ventricular (RV) pressures but also PVR, thus providing indirect evidence for reverse remodeling in the pulmonary vasculature. Using an established rat model of PH-LHD due to left heart failure triggered by pressure overload with subsequent development of PH, a model is developed to study the molecular and cellular mechanisms of this physiological reverse remodeling process. Specifically, an aortic debanding surgery was performed, which resulted in reverse remodeling of the LV myocardium and its unloading. In parallel, complete normalization of RV systolic pressure and significant but incomplete reversal of RV hypertrophy was detectable. This model may present a valuable tool to study the mechanisms of physiological reverse remodeling in the pulmonary circulation and the RV, aiming to develop therapeutic strategies for treating PH-LHD and other forms of PH.


Assuntos
Insuficiência Cardíaca , Hipertensão Pulmonar , Animais , Humanos , Hipertensão Pulmonar/etiologia , Artéria Pulmonar/cirurgia , Circulação Pulmonar/fisiologia , Ratos , Remodelação Vascular
5.
Elife ; 102021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33620318

RESUMO

To assemble a brain, differentiating neurons must make proper connections and establish specialized brain compartments. Abnormal levels of cell adhesion molecules disrupt these processes. Dystroglycan (Dg) is a major non-integrin cell adhesion receptor, deregulation of which is associated with dramatic neuroanatomical defects such as lissencephaly type II or cobblestone brain. The previously established Drosophila model for cobblestone lissencephaly was used to understand how Dg is regulated in the brain. During development, Dg has a spatiotemporally dynamic expression pattern, fine-tuning of which is crucial for accurate brain assembly. In addition, mass spectrometry analyses identified numerous components associated with Dg in neurons, including several proteins of the exocyst complex. Data show that exocyst-based membrane trafficking of Dg allows its distinct expression pattern, essential for proper brain morphogenesis. Further studies of the Dg neuronal interactome will allow identification of new factors involved in the development of dystroglycanopathies and advance disease diagnostics in humans.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Distroglicanas/genética , Lisencefalia/genética , Animais , Modelos Animais de Doenças , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Distroglicanas/metabolismo , Larva/genética , Larva/crescimento & desenvolvimento , Neurônios/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo
6.
BMC Med ; 18(1): 8, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31959160

RESUMO

BACKGROUND: Dystroglycanopathies are a group of inherited disorders characterized by vast clinical and genetic heterogeneity and caused by abnormal functioning of the ECM receptor dystroglycan (Dg). Remarkably, among many cases of diagnosed dystroglycanopathies, only a small fraction can be linked directly to mutations in Dg or its regulatory enzymes, implying the involvement of other, not-yet-characterized, Dg-regulating factors. To advance disease diagnostics and develop new treatment strategies, new approaches to find dystroglycanopathy-related factors should be considered. The Dg complex is highly evolutionarily conserved; therefore, model genetic organisms provide excellent systems to address this challenge. In particular, Drosophila is amenable to experiments not feasible in any other system, allowing original insights about the functional interactors of the Dg complex. METHODS: To identify new players contributing to dystroglycanopathies, we used Drosophila as a genetic muscular dystrophy model. Using mass spectrometry, we searched for muscle-specific Dg interactors. Next, in silico analyses allowed us to determine their association with diseases and pathological conditions in humans. Using immunohistochemical, biochemical, and genetic interaction approaches followed by the detailed analysis of the muscle tissue architecture, we verified Dg interaction with some of the discovered factors. Analyses of mouse muscles and myocytes were used to test if interactions are conserved in vertebrates. RESULTS: The muscle-specific Dg complexome revealed novel components that influence the efficiency of Dg function in the muscles. We identified the closest human homologs for Dg-interacting partners, determined their significant enrichment in disease-associations, and verified some of the newly identified Dg interactions. We found that Dg associates with two components of the mechanosignaling Hippo pathway: the WW domain-containing proteins Kibra and Yorkie. Importantly, this conserved interaction manages adult muscle size and integrity. CONCLUSIONS: The results presented in this study provide a new list of muscle-specific Dg interactors, further analysis of which could aid not only in the diagnosis of muscular dystrophies, but also in the development of new therapeutics. To regulate muscle fitness during aging and disease, Dg associates with Kibra and Yorkie and acts as a transmembrane Hippo signaling receptor that transmits extracellular information to intracellular signaling cascades, regulating muscle gene expression.


Assuntos
Proteínas de Drosophila/metabolismo , Distroglicanas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Atrofia Muscular/metabolismo , Distrofias Musculares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Envelhecimento/metabolismo , Animais , Modelos Animais de Doenças , Drosophila , Distroglicanas/genética , Feminino , Masculino , Espectrometria de Massas , Camundongos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/patologia , Distrofias Musculares/genética , Distrofias Musculares/patologia , Mutação , Mapas de Interação de Proteínas
8.
J Cell Sci ; 131(4)2018 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-29444950

RESUMO

Stress can be temporary or chronic, and mild or acute. Depending on its extent and severity, cells either alter their metabolism, and adopt a new state, or die. Fluctuations in environmental conditions occur frequently, and such stress disturbs cellular homeostasis, but in general, stresses are reversible and last only a short time. There is increasing evidence that regulation of gene expression in response to temporal stress happens post-transcriptionally in specialized subcellular membrane-less compartments called ribonucleoprotein (RNP) granules. RNP granules assemble through a concentration-dependent liquid-liquid phase separation of RNA-binding proteins that contain low-complexity sequence domains (LCDs). Interestingly, many factors that regulate microRNA (miRNA) biogenesis and alternative splicing are RNA-binding proteins that contain LCDs and localize to stress-induced liquid-like compartments. Consequently, gene silencing through miRNAs and alternative splicing of pre-mRNAs are emerging as crucial post-transcriptional mechanisms that function on a genome-wide scale to regulate the cellular stress response. In this Review, we describe the interplay between these two post-transcriptional processes that occur in liquid-like compartments as an adaptive cellular response to stress.


Assuntos
Processamento Alternativo/genética , MicroRNAs/genética , Ribonucleoproteínas/genética , Estresse Fisiológico/genética , Regulação da Expressão Gênica/genética , Inativação Gênica , Precursores de RNA/genética , Processamento Pós-Transcricional do RNA/genética , Proteínas de Ligação a RNA/genética
9.
Nat Commun ; 9(1): 312, 2018 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-29358748

RESUMO

Upon stress, profound post-transcriptional adjustments of gene expression occur in spatially restricted, subcellular, membraneless compartments, or ribonucleoprotein (RNP) granules, which are formed by liquid phase separation of RNA-binding proteins with low complexity sequence domains (LCDs). Here, we show that Rbfox1 is an LCD-containing protein that aggregates into liquid droplets and amyloid-like fibers and promiscuously joins different nuclear and cytoplasmic RNP granules. Using Drosophila oogenesis as an in vivo system for stress response, we demonstrate a mechanism by which Rbfox1 promotes cell survival. The stress-dependent miRNA miR-980 acts to buffer Rbfox1 levels, since it targets only those Rbfox1 transcripts that contain extended 3'UTRs. Reduced miR-980 expression during stress leads to increased Rbfox1 levels, widespread formation of various RNP granules, and increased cell viability. We show that human RBFOX proteins also contain multiple LCDs and form membraneless compartments, suggesting that the RNP granule-linked control of cellular adaptive responses may contribute to a wide range of RBFOX-associated pathologies in humans.


Assuntos
Proteínas de Drosophila/genética , MicroRNAs/genética , Oócitos/metabolismo , Proteínas de Ligação a RNA/genética , Ribonucleoproteínas/genética , Estresse Fisiológico/genética , Adaptação Fisiológica , Animais , Sobrevivência Celular , Reprogramação Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , MicroRNAs/metabolismo , Mutação , Neurônios/citologia , Neurônios/metabolismo , Oócitos/citologia , Oogênese/genética , Ovário/citologia , Ovário/metabolismo , Cultura Primária de Células , Domínios Proteicos , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/metabolismo
10.
BMC Neurosci ; 17(1): 72, 2016 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-27829368

RESUMO

BACKGROUND: The AP-2 transcription factor APTF-1 is crucially required for developmentally controlled sleep behavior in Caenorhabditis elegans larvae. Its human ortholog, TFAP-2beta, causes Char disease and has also been linked to sleep disorders. These data suggest that AP-2 transcription factors may be highly conserved regulators of various types of sleep behavior. Here, we tested the idea that AP-2 controls adult sleep in Drosophila. RESULTS: Drosophila has one AP-2 ortholog called TfAP-2, which is essential for viability. To investigate its potential role in sleep behavior and neural development, we specifically downregulated TfAP-2 in the nervous system. We found that neuronal TfAP-2 knockdown almost completely abolished night sleep but did not affect day sleep. TfAP-2 insufficiency affected nervous system development. Conditional TfAP-2 knockdown in the adult also produced a modest sleep phenotype, suggesting that TfAP-2 acts both in larval as well as in differentiated neurons. CONCLUSIONS: Thus, our results show that AP-2 transcription factors are highly conserved regulators of development and sleep.


Assuntos
Proteínas de Drosophila/metabolismo , Sono/fisiologia , Fator de Transcrição AP-2/metabolismo , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Drosophila/genética , Drosophila melanogaster , Técnicas de Silenciamento de Genes , Imuno-Histoquímica , Masculino , Neurônios/metabolismo , Neurônios/patologia , Fotoperíodo , Filogenia , Reação em Cadeia da Polimerase em Tempo Real , Fator de Transcrição AP-2/genética , Gravação em Vídeo
11.
J Vis Exp ; (88): e51681, 2014 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-24998593

RESUMO

Metabolic disorders are a frequent problem affecting human health. Therefore, understanding the mechanisms that regulate metabolism is a crucial scientific task. Many disease causing genes in humans have a fly homologue, making Drosophila a good model to study signaling pathways involved in the development of different disorders. Additionally, the tractability of Drosophila simplifies genetic screens to aid in identifying novel therapeutic targets that may regulate metabolism. In order to perform such a screen a simple and fast method to identify changes in the metabolic state of flies is necessary. In general, carbon dioxide production is a good indicator of substrate oxidation and energy expenditure providing information about metabolic state. In this protocol we introduce a simple method to measure CO2 output from flies. This technique can potentially aid in the identification of genetic perturbations affecting metabolic rate.


Assuntos
Dióxido de Carbono/metabolismo , Drosophila melanogaster/metabolismo , Animais , Dióxido de Carbono/análise , Drosophila melanogaster/genética , Metabolismo Energético , Masculino , Modelos Animais , Oxirredução , Mecânica Respiratória/fisiologia
12.
Fly (Austin) ; 7(3): 173-83, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23839338

RESUMO

The generation of neuronal cell diversity is controlled by interdependent mechanisms, including cell intrinsic programs and environmental cues. During development, the astonishing variety of neurons is originated according to a precise timetable that is managed by a complex network of genes specifying individual types of neurons. Different neurons express specific sets of transcription factors, and they can be recognized by morphological characteristics and spatial localization, but, most importantly, they connect to each other and form functional units in a stereotyped fashion. This connectivity depends, mostly, on selective cell adhesion that is strictly regulated. While intrinsic factors specifying neuronal temporal identity have been extensively studied, an extrinsic temporal factor controlling neuronal temporal identity switch has not been shown. Our data demonstrate that pulses of steroid hormone act as a temporal cue to fine-tune neuronal cell differentiation. Here we also provide evidence that extrinsic JAK/STAT cytokine signaling acts as a spatial code in the process. Particularly, in Drosophila mushroom bodies, neuronal identity transition is controlled by steroid-dependent microRNAs that regulate spatially distributed cytokine-dependent signaling factors that in turn modulate cell adhesion. A new era of neuronal plasticity assessment via managing external temporal cues such as hormones and cytokines that specify individual types of neurons might open new possibilities for brain regenerative therapeutics.


Assuntos
Diferenciação Celular/fisiologia , Drosophila/crescimento & desenvolvimento , Hormônios Esteroides Gonadais/metabolismo , MicroRNAs/fisiologia , Corpos Pedunculados/crescimento & desenvolvimento , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Animais
13.
EMBO J ; 31(24): 4511-23, 2012 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-23160410

RESUMO

Mammalian neuronal stem cells produce multiple neuron types in the course of an individual's development. Similarly, neuronal progenitors in the Drosophila brain generate different types of closely related neurons that are born at specific time points during development. We found that in the post-embryonic Drosophila brain, steroid hormones act as temporal cues that specify the cell fate of mushroom body (MB) neuroblast progeny. Chronological regulation of neurogenesis is subsequently mediated by the microRNA (miRNA) let-7, absence of which causes learning impairment due to morphological MB defects. The miRNA let-7 is required to regulate the timing of α'/ß' to α/ß neuronal identity transition by targeting the transcription factor Abrupt. At a cellular level, the ecdysone-let-7-Ab signalling pathway controls the expression levels of the cell adhesion molecule Fasciclin II in developing neurons that ultimately influences their differentiation. Our data propose a novel role for miRNAs as transducers between chronologically regulated developmental signalling and physical cell adhesion.


Assuntos
Diferenciação Celular/fisiologia , Drosophila/crescimento & desenvolvimento , Hormônios Esteroides Gonadais/metabolismo , MicroRNAs/fisiologia , Corpos Pedunculados/crescimento & desenvolvimento , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Animais , Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas de Drosophila/metabolismo , Ecdisona/metabolismo , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Imuno-Histoquímica , Hibridização In Situ , MicroRNAs/metabolismo , Corpos Pedunculados/citologia , Proteínas Nucleares/metabolismo , Transdução de Sinais/fisiologia
14.
BMC Cell Biol ; 13: 26, 2012 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-23107381

RESUMO

BACKGROUND: The Dystrophin Glycoprotein Complex (DGC) is at the center of significant inheritable diseases, such as muscular dystrophies that can be fatal and impair neuronal function in addition to muscle degeneration. Recent evidence has shown that it can control cellular homeostasis and work via Dystrophin signaling to regulate microRNA gene expression which implies that disease phenotypes hide an entourage of regulatory and homeostatic anomalies. Uncovering these hidden processes could shed new light on the importance of proper DGC function for an organism's overall welfare and bring forth new ideas for treatments. RESULTS: To better understand a role for the DGC in these processes, we used the genetically advantageous Drosophila muscular dystrophy model to conduct a whole animal microarray screen. Since we have recently found that dystrophic symptoms can be caused by stress even in wild type animals and are enhanced in mutants, we screened stressed animals for microRNA misregulation as well. We were able to define microRNAs misregulated due to stress and/or dystrophy. Our results support the hypothesis that there is a Dystrophin and Dystroglycan dependent circuitry of processes linking stress response, dystrophic conditions and cellular signaling and that microRNAs play an important role in this network. Verification of a subset of our results was conducted via q-PCR and revealed that miR-956, miR-980 and miR-252 are regulated via a Dystroglycan-Dystrophin-Syntrophin dependent pathway. CONCLUSIONS: The results presented in this study support the hypothesis that there is a Dystrophin and Dystroglycan dependent circuitry of processes that includes regulation of microRNAs. Dystrophin signaling has already been found to occur in mammalian musculature; however, our data reveals that this regulation is evolutionarily conserved and also present in at least neuronal tissues. Our data imply that Dystroglycan-Dystrophin-Syntrophin signaling through control of multiple microRNAs is involved in highly managed regulation of gene expression required to adapt cellular homeostasis that is compromised under stress and dystrophic conditions.


Assuntos
Proteínas de Drosophila/metabolismo , Distroglicanas/metabolismo , Proteínas Associadas à Distrofina/metabolismo , Distrofina/metabolismo , MicroRNAs/metabolismo , Animais , Drosophila/metabolismo , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Transdução de Sinais
15.
BMC Neurosci ; 12: 93, 2011 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-21943192

RESUMO

BACKGROUND: The Dystrophin Glycoprotein Complex (DGC) is a large multi-component complex that is well known for its function in muscle tissue. When the main components of the DGC, Dystrophin (Dys) and Dystroglycan (Dg) are affected cognitive impairment and mental retardation in addition to muscle degeneration can occur. Previously we performed an array of genetic screens using a Drosophila model for muscular dystrophy in order to find novel DGC interactors aiming to elucidate the signaling role(s) in which the complex is involved. Since the function of the DGC in the brain and nervous system has not been fully defined, we have here continued to analyze the DGC modifiers' function in the developing Drosophila brain and eye. RESULTS: Given that disruption of Dys and Dg leads to improper photoreceptor axon projections into the lamina and eye neuron elongation defects during development, we have determined the function of previously screened components and their genetic interaction with the DGC in this tissue. Our study first found that mutations in chif, CG34400, Nrk, Lis1, capt and Cam cause improper axon path-finding and loss of SP2353, Grh, Nrk, capt, CG34400, vimar, Lis1 and Cam cause shortened rhabdomere lengths. We determined that Nrk, mbl, capt and Cam genetically interact with Dys and/or Dg in these processes. It is notable that most of the neuronal DGC interacting components encountered are involved in regulation of actin dynamics. CONCLUSIONS: Our data indicate possible DGC involvement in the process of cytoskeletal remodeling in neurons. The identification of new components that interact with the DGC not only helps to dissect the mechanism of axon guidance and eye neuron differentiation but also provides a great opportunity for understanding the signaling mechanisms by which the cell surface receptor Dg communicates via Dys with the actin cytoskeleton.


Assuntos
Axônios/metabolismo , Proteínas de Drosophila/metabolismo , Distroglicanas/metabolismo , Distrofina/metabolismo , Células Fotorreceptoras de Invertebrados/metabolismo , Animais , Axônios/patologia , Axônios/fisiologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Drosophila melanogaster , Distroglicanas/genética , Distroglicanas/fisiologia , Distrofina/genética , Distrofina/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Mutação , Células Fotorreceptoras de Invertebrados/patologia , Células Fotorreceptoras de Invertebrados/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
16.
Dev Biol ; 352(2): 228-42, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21256839

RESUMO

In Drosophila, like in humans, Dystrophin Glycoprotein Complex (DGC) deficiencies cause a life span shortening disease, associated with muscle dysfunction. We performed the first in vivo genetic interaction screen in ageing dystrophic muscles and identified genes that have not been shown before to have a role in the development of muscular dystrophy and interact with dystrophin and/or dystroglycan. Mutations in many of the found interacting genes cause age-dependent morphological and heat-induced physiological defects in muscles, suggesting their importance in the tissue. Majority of them is phylogenetically conserved and implicated in human disorders, mainly tumors and myopathies. Functionally they can be divided into three main categories: proteins involved in communication between muscle and neuron, and interestingly, in mechanical and cellular stress response pathways. Our data show that stress induces muscle degeneration and accelerates age-dependent muscular dystrophy. Dystrophic muscles are already compromised; and as a consequence they are less adaptive and more sensitive to energetic stress and to changes in the ambient temperature. However, only dystroglycan, but not dystrophin deficiency causes extreme myodegeneration induced by energetic stress suggesting that dystroglycan might be a component of the low-energy pathway and act as a transducer of energetic stress in normal and dystrophic muscles.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/genética , Drosophila/metabolismo , Distroglicanas/genética , Distroglicanas/metabolismo , Distrofina/genética , Distrofina/metabolismo , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Estresse Fisiológico , Animais , Sequência de Bases , Primers do DNA/genética , Modelos Animais de Doenças , Distroglicanas/antagonistas & inibidores , Distroglicanas/deficiência , Distrofina/antagonistas & inibidores , Distrofina/deficiência , Feminino , Genes de Insetos , Humanos , Masculino , Células Musculares/metabolismo , Distrofia Muscular Animal/etiologia , Mutação , Interferência de RNA , Transdução de Sinais
17.
Sci Rep ; 1: 47, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22355566

RESUMO

In humans, mutations in the Dystrophin Glycoprotein Complex (DGC) cause muscular dystrophies (MDs) that are associated with muscle loss, seizures and brain abnormalities leading to early death. Using Drosophila as a model to study MD we have found that loss of Dystrophin (Dys) during development leads to heat-sensitive abnormal muscle contractions that are repressed by mutations in Dys's binding partner, Dystroglycan (Dg). Hyperthermic seizures are independent from dystrophic muscle degeneration and rely on neurotransmission, which suggests involvement of the DGC in muscle-neuron communication. Additionally, reduction of the Ca(2+) regulator, Calmodulin or Ca(2+) channel blockage rescues the seizing phenotype, pointing to Ca(2+) mis-regulation in dystrophic muscles. Also, Dys and Dg mutants have antagonistically abnormal cellular levels of ROS, suggesting that the DGC has a function in regulation of muscle cell homeostasis. These data show that muscles deficient for Dys are predisposed to hypercontraction that may result from abnormal neuromuscular junction signaling.


Assuntos
Sinalização do Cálcio , Febre/fisiopatologia , Homeostase , Contração Muscular , Rigidez Muscular/fisiopatologia , Músculo Esquelético/fisiopatologia , Distrofias Musculares/fisiopatologia , Animais , Drosophila , Febre/complicações , Humanos , Rigidez Muscular/complicações , Convulsões
18.
J Vis Exp ; (46)2010 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-21206479

RESUMO

The molecular characterization of muscular dystrophies and myopathies in humans has revealed the complexity of muscle disease and genetic analysis of muscle specification, formation and function in model systems has provided valuable insight into muscle physiology. Therefore, identifying and characterizing molecular mechanisms that underlie muscle damage is critical. The structure of adult Drosophila multi-fiber muscles resemble vertebrate striated muscles (1) and the genetic tractability of Drosophila has made it a great system to analyze dystrophic muscle morphology and characterize the processes affecting muscular function in ageing adult flies (2). Here we present the histological technique for preparing paraffin-embedded and frozen sections of Drosophila thoracic muscles. These preparations allow for the tissue to be stained with classical histological stains and labeled with protein detecting dyes, and specifically cryosections are ideal for immunohistochemical detection of proteins in intact muscles. This allows for analysis of muscle tissue structure, identification of morphological defects, and detection of the expression pattern for muscle/neuron-specific proteins in Drosophila adult muscles. These techniques can also be slightly modified for sectioning of other body parts.


Assuntos
Drosophila/anatomia & histologia , Músculo Esquelético/anatomia & histologia , Inclusão em Parafina/métodos , Animais , Drosophila/fisiologia , Músculo Esquelético/fisiologia , Coloração e Rotulagem/métodos , Tórax/anatomia & histologia
19.
PLoS One ; 3(6): e2418, 2008 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-18545683

RESUMO

The Dystroglycan-Dystrophin (Dg-Dys) complex has a capacity to transmit information from the extracellular matrix to the cytoskeleton inside the cell. It is proposed that this interaction is under tight regulation; however the signaling/regulatory components of Dg-Dys complex remain elusive. Understanding the regulation of the complex is critical since defects in this complex cause muscular dystrophy in humans. To reveal new regulators of the Dg-Dys complex, we used a model organism Drosophila melanogaster and performed genetic interaction screens to identify modifiers of Dg and Dys mutants in Drosophila wing veins. These mutant screens revealed that the Dg-Dys complex interacts with genes involved in muscle function and components of Notch, TGF-beta and EGFR signaling pathways. In addition, components of pathways that are required for cellular and/or axonal migration through cytoskeletal regulation, such as Semaphorin-Plexin, Frazzled-Netrin and Slit-Robo pathways show interactions with Dys and/or Dg. These data suggest that the Dg-Dys complex and the other pathways regulating extracellular information transfer to the cytoskeletal dynamics are more intercalated than previously thought.


Assuntos
Drosophila melanogaster/genética , Distroglicanas/metabolismo , Distrofina/metabolismo , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Receptores ErbB/metabolismo , Mutação , Receptores Notch/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
20.
J Biol Chem ; 282(20): 15159-69, 2007 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-17355978

RESUMO

The conserved dystroglycan-dystrophin (Dg.Dys) complex connects the extracellular matrix to the cytoskeleton. In humans as well as Drosophila, perturbation of this complex results in muscular dystrophies and brain malformations and in some cases cellular polarity defects. However, the regulation of the Dg.Dys complex is poorly understood in any cell type. We now find that in loss-of-function and overexpression studies more than half (34 residues) of the Dg proline-rich conserved C-terminal regions can be truncated without significantly compromising its function in regulating cellular polarity in Drosophila. Notably, the truncation eliminates the WW domain binding motif at the very C terminus of the protein thought to mediate interactions with dystrophin, suggesting that a second, internal WW binding motif can also mediate this interaction. We confirm this hypothesis by using a sensitive fluorescence polarization assay to show that both WW domain binding sites of Dg bind to Dys in humans (K(d) = 7.6 and 81 microM, respectively) and Drosophila (K(d) = 16 and 46 microM, respectively). In contrast to the large deletion mentioned above, a single proline to an alanine point mutation within a predicted Src homology 3 domain (SH3) binding site abolishes Dg function in cellular polarity. This suggests that an SH3-containing protein, which has yet to be identified, functionally interacts with Dg.


Assuntos
Polaridade Celular/fisiologia , Proteínas de Drosophila/metabolismo , Distroglicanas/metabolismo , Distrofina/metabolismo , Domínios de Homologia de src , Motivos de Aminoácidos , Sequência de Aminoácidos/genética , Substituição de Aminoácidos , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster , Distroglicanas/genética , Distrofina/genética , Humanos , Mutação Puntual , Ligação Proteica/genética , Deleção de Sequência , Homologia de Sequência de Aminoácidos , Domínios de Homologia de src/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...