Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Development ; 150(20)2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-37795747

RESUMEN

Obesity is linked to reduced fertility in various species, from Drosophila to humans. Considering that obesity is often induced by changes in diet or eating behavior, it remains unclear whether obesity, diet, or both reduce fertility. Here, we show that Drosophila females on a high-sugar diet become rapidly obese and less fertile as a result of increased death of early germline cysts and vitellogenic egg chambers (or follicles). They also have high glycogen, glucose and trehalose levels and develop insulin resistance in their fat bodies (but not ovaries). By contrast, females with adipocyte-specific knockdown of the anti-obesity genes brummer or adipose are obese but have normal fertility. Remarkably, females on a high-sugar diet supplemented with a separate source of water have mostly normal fertility and glucose levels, despite persistent obesity, high glycogen and trehalose levels, and fat body insulin resistance. These findings demonstrate that a high-sugar diet affects specific processes in oogenesis independently of insulin resistance, that high glucose levels correlate with reduced fertility on a high-sugar diet, and that obesity alone does not impair fertility.


Asunto(s)
Drosophila melanogaster , Resistencia a la Insulina , Animales , Humanos , Femenino , Drosophila melanogaster/genética , Trehalosa , Obesidad/etiología , Dieta , Drosophila , Fertilidad , Glucosa , Glucógeno
2.
Development ; 149(5)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35156684

RESUMEN

Despite their medical and economic relevance, it remains largely unknown how suboptimal temperatures affect adult insect reproduction. Here, we report an in-depth analysis of how chronic adult exposure to suboptimal temperatures affects oogenesis using the model insect Drosophila melanogaster. In adult females maintained at 18°C (cold) or 29°C (warm), relative to females at the 25°C control temperature, egg production was reduced through distinct cellular mechanisms. Chronic 18°C exposure improved germline stem cell maintenance, survival of early germline cysts and oocyte quality, but reduced follicle growth with no obvious effect on vitellogenesis. By contrast, in females at 29°C, germline stem cell numbers and follicle growth were similar to those at 25°C, while early germline cyst death and degeneration of vitellogenic follicles were markedly increased and oocyte quality plummeted over time. Finally, we also show that these effects are largely independent of diet, male factors or canonical temperature sensors. These findings are relevant not only to cold-blooded organisms, which have limited thermoregulation, but also potentially to warm-blooded organisms, which are susceptible to hypothermia, heatstroke and fever.


Asunto(s)
Linaje de la Célula/fisiología , Drosophila melanogaster/fisiología , Células Germinativas/fisiología , Oogénesis/fisiología , Células Madre/fisiología , Animales , Frío , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Masculino , Oocitos/fisiología , Folículo Ovárico/fisiología , Ovario/fisiología , Transducción de Señal/fisiología , Vitelogénesis/fisiología
3.
Development ; 148(5)2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33547134

RESUMEN

The conserved nuclear receptor superfamily has crucial roles in many processes, including reproduction. Nuclear receptors with known roles in oogenesis have been studied mostly in the context of their ovary-intrinsic requirement. Recent studies in Drosophila, however, have begun to reveal new roles of nuclear receptor signaling in peripheral tissues in controlling reproduction. Here, we identified Hormone receptor 4 (Hr4) as an oogenesis regulator required in the ovary and muscles. Global Hr4 knockdown leads to increased germline stem cell (GSC) loss, reduced GSC proliferation, early germline cyst death, slowed follicle growth and vitellogenic follicle degeneration. Tissue-specific knockdown experiments uncovered ovary-intrinsic and peripheral tissue requirements for Hr4 In the ovary, Hr4 is required in the niche for GSC proliferation and in the germline for GSC maintenance. Hr4 functions in muscles to promote GSC maintenance and follicle growth. The specific tissues that require Hr4 for survival of early germline cysts and vitellogenic follicles remain unidentified. These results add to the few examples of muscles controlling gametogenesis and expand our understanding of the complexity of nuclear receptor regulation of various aspects of oogenesis.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Músculos/metabolismo , Oogénesis/genética , Ovario/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Animales Modificados Genéticamente/metabolismo , Proliferación Celular , Drosophila/crecimiento & desarrollo , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Femenino , Células Germinativas/citología , Células Germinativas/metabolismo , Músculos/citología , Folículo Ovárico/crecimiento & desarrollo , Folículo Ovárico/metabolismo , Ovario/citología , Óvulo/crecimiento & desarrollo , Óvulo/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/genética
4.
Development ; 147(11)2020 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-32540896

RESUMEN

Developmental biologists have frequently pushed the frontiers of modern biomedical research. From the discovery and characterization of novel signal transduction pathways to exploring the molecular underpinnings of genetic inheritance, transcription, the cell cycle, cell death and stem cell biology, studies of metazoan development have historically opened new fields of study and consistently revealed previously unforeseen avenues of clinical therapies. From this perspective, it is not surprising that our community is now an integral part of the current renaissance in metabolic research. Amidst the global rise in metabolic syndrome, the discovery of novel signaling roles for metabolites, and the increasing links between altered metabolism and many human diseases, we as developmental biologists can contribute skills and expertise that are uniquely suited for investigating the mechanisms underpinning human metabolic health and disease. Here, we summarize the opportunities and challenges that our community faces, and discuss how developmental biologists can make unique and valuable contributions to the field of metabolism and physiology.


Asunto(s)
Biología Evolutiva , Enfermedades Metabólicas/metabolismo , Animales , Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Humanos , Enfermedades Metabólicas/patología , Neoplasias/metabolismo , Neoplasias/patología , Oogénesis , Erizos de Mar/crecimiento & desarrollo , Erizos de Mar/metabolismo , Transducción de Señal
5.
Dev Biol ; 456(2): 179-189, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31470019

RESUMEN

Reproduction is intimately linked to the physiology of an organism. Nuclear receptors are widely expressed transcription factors that mediate the effects of many circulating molecules on physiology and reproduction. While multiple studies have focused on the roles of nuclear receptors intrinsically in the ovary, it remains largely unknown how the actions of nuclear receptors in peripheral tissues influence oogenesis. We identified the nuclear receptor encoded by svp as a novel regulator of oogenesis in adult Drosophila. Global somatic knockdown of svp reduces egg production by increasing GSC loss, death of early germline cysts, and degeneration of vitellogenic follicles. Tissue-specific knockdown experiments revealed that svp remotely controls these different steps of oogenesis through separate mechanisms involving distinct tissues. Specifically, adipocyte-specific svp knockdown impairs GSC maintenance and early germline cyst survival, whereas oenocyte-specific svp knockdown increases the death of vitellogenic follicles without any effects on GSCs or early cysts. These results illustrate that nuclear receptors can control reproduction through a variety of mechanisms involving peripheral tissues.


Asunto(s)
Adipocitos/metabolismo , Proteínas de Unión al ADN/metabolismo , Oogénesis/fisiología , Receptores de Esteroides/metabolismo , Adipocitos/fisiología , Animales , Proteínas de Unión al ADN/fisiología , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Células Germinativas/metabolismo , Ovario/metabolismo , Receptores Citoplasmáticos y Nucleares , Receptores de Esteroides/fisiología , Transducción de Señal/fisiología , Factores de Transcripción
6.
J Infect Dis ; 219(3): 480-488, 2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30165577

RESUMEN

Background: Cutaneous leishmaniasis (CL) is characterized by an exaggerated inflammatory response. During pregnancy there is a decreased inflammatory response, and we have shown that pregnant women with CL develop exuberant lesions. Methods: Cytokine production by peripheral blood mononuclear cells and the frequency of cells expressing cytokines in lesions from pregnant and nonpregnant women with CL were evaluated. Results: We observed that CL lesions from pregnant women displayed a more intense cellular infiltrate, associated with an increase in neutrophils and CD4+ cells. While no difference was observed regarding the number of interferon-gamma (IFN-γ)+ cells in lesions from pregnant compared to nonpregnant women with CL, interleukin-10 (IL-10) and IL-4 expression were approximately 3-times higher in lesions in pregnant women. Main sources of IL-4 and IL-10 were CD4+ and CD68+ cells, respectively. Expression of IL-4, but not IFN-γ or IL-10, was positively correlated with the intensity of inflammatory infiltrate in lesions from pregnant women. Conclusions: These results provide evidence of an IL-4-mediated pathology in Leishmania braziliensis-infected pregnant women. These differences in lesion pathogenesis in pregnant and nonpregnant women may open possibilities for new therapies for CL treatment during pregnancy, which are currently lacking.


Asunto(s)
Leishmania braziliensis/inmunología , Leishmaniasis Cutánea/inmunología , Células Th2/inmunología , Adolescente , Adulto , Linfocitos T CD4-Positivos/inmunología , Citocinas/metabolismo , Femenino , Humanos , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Leishmaniasis Cutánea/patología , Embarazo , Piel/patología , Adulto Joven
7.
Dev Biol ; 440(1): 31-39, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29729259

RESUMEN

Tissue-specific stem cells are tied to the nutritional and physiological environment of adult organisms. Adipocytes have key endocrine and nutrient-sensing roles and have emerged as major players in relaying dietary information to regulate other organs. For example, previous studies in Drosophila melanogaster revealed that amino acid sensing as well as diet-dependent metabolic pathways function in adipocytes to influence the maintenance of female germline stem cells (GSCs). How nutrient-sensing pathways acting within adipocytes influence adult stem cell lineages, however, is just beginning to be elucidated. Here, we report that insulin/insulin-like growth factor signaling in adipocytes promotes GSC maintenance, early germline cyst survival, and vitellogenesis. Further, adipocytes use distinct mechanisms downstream of insulin receptor activation to control these aspects of oogenesis, all of which are independent of FOXO. We find that GSC maintenance is modulated by Akt1 through GSK-3ß, early germline cyst survival is downstream of adipocyte Akt1 but independent of GSK-3ß, and vitellogenesis is regulated through an Akt1-independent pathway in adipocytes. These results indicate that, in addition to employing different types of nutrient sensing, adipocytes can use distinct axes of a single nutrient-sensing pathway to regulate multiple stages of the GSC lineage in the ovary.


Asunto(s)
Adipocitos/fisiología , Células Madre Germinales Adultas/fisiología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Adipocitos/metabolismo , Células Madre Germinales Adultas/metabolismo , Animales , Recuento de Células , Proliferación Celular , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Femenino , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/fisiología , Células Germinativas/citología , Glucógeno Sintasa Quinasa 3 beta/fisiología , Insulina/metabolismo , Masculino , Oogénesis/fisiología , Ovario/citología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Células Madre/citología
8.
Eur J Anaesthesiol ; 36(7): 502-508, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30985540

RESUMEN

BACKGROUND: Magnesium sulphate is an important adjuvant drug in multimodal anaesthesia. In combination with rocuronium it can enhance neuromuscular blockade (NMB). Limited data exist concerning the effect of magnesium sulphate on the duration of deep or intense NMB and the period of no response. OBJECTIVE(S): To determine the role of magnesium sulphate on the duration of rocuronium-induced deep and intense NMB, and the period of no response to nerve stimulation. DESIGN: A randomised controlled trial. SETTING: A public tertiary care hospital, Rio de Janeiro, Brazil, from February 2017 to March 2018. PATIENTS: All patients between 18 and 65 years of age scheduled to undergo elective otorhinolaryngological surgery, with a BMI between 18.5 and 24.9 kg m and an American Society of Anesthesiologists physical status classification of I or II. INTERVENTION(S): Before induction of anaesthesia 60 patients were pretreated with an intravenous infusion of either 100 ml 0.9% saline (saline group), or 60 mg kg magnesium sulphate (magnesium group). After loss of consciousness, a bolus of rocuronium (0.6 mg kg) was administered. Neuromuscular function was measured by TOF-Watch SX monitor. MAIN OUTCOME MEASURES: The primary and secondary outcomes were the duration of the period of no response to nerve stimulation and intense and deep NMB, respectively. An additional outcome was the NMB onset time. RESULTS: Median [IQR] durations of deep NMB were 20.3 [12.0 to 35.4] and 18.3 [11.2 to 26.3] min in the magnesium and saline groups, respectively (P = 0.18). Median durations of intense NMB were 21.7 [0.0 to 32.2] min and 0.0 [0.0 to 6.2] min (P = 0.001) in the magnesium and saline groups, respectively. Median durations of the period of no response were 40.8 [51.4 to 36.0] min and 28.0 [21.9 to 31.6] min (P = 0.0001) in the magnesium and saline groups, respectively. CONCLUSION: Magnesium sulphate increased both the duration of intense NMB and the period of no response. The duration of deep NMB was similar in the magnesium sulphate group and saline group. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT02989272.


Asunto(s)
Anestésicos/administración & dosificación , Sulfato de Magnesio/administración & dosificación , Bloqueo Neuromuscular/métodos , Rocuronio/administración & dosificación , Adolescente , Adulto , Brasil , Método Doble Ciego , Femenino , Humanos , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Fármacos Neuromusculares no Despolarizantes/administración & dosificación , Procedimientos Quirúrgicos Otorrinolaringológicos/métodos , Factores de Tiempo , Adulto Joven
9.
Dev Biol ; 420(1): 90-99, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27729213

RESUMEN

Multiple aspects of organismal physiology influence the number and activity of stem cells and their progeny, including nutritional status. Previous studies demonstrated that Drosophila germline stem cells (GSCs), follicle stem cells (FSCs), and their progeny sense and respond to diet via complex mechanisms involving many systemic and local signals. AMP-activated protein kinase, or AMPK, is a highly conserved regulator of energy homeostasis known to be activated under low cellular energy conditions; however, its role in the ovarian response to diet has not been investigated. Here, we describe nutrient-dependent and -independent requirements for AMPK in Drosophila oogenesis. We found that AMPK is cell autonomously required for the slow down in GSC and follicle cell proliferation that occurs on a poor diet. Similarly, AMPK activity is necessary in the germline for the degeneration of vitellogenic stages in response to nutrient deprivation. In contrast, AMPK activity is not required within the germline to modulate its growth. Instead, AMPK acts in follicle cells to negatively regulate their growth and proliferation, thereby indirectly limiting the size of the underlying germline cyst within developing follicles. Paradoxically, AMPK is required for GSC maintenance in well-fed flies (when AMPK activity is presumably at its lowest), suggesting potentially important roles for basal AMPK activity in specific cell types. Finally, we identified a nutrient-independent, developmental role for AMPK in cyst encapsulation by follicle cells. These results uncover specific AMPK requirements in multiple cell types in the ovary and suggest that AMPK can function outside of its canonical nutrient-sensing role in specific developmental contexts.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Dieta , Drosophila melanogaster/metabolismo , Oogénesis , Animales , Proliferación Celular , Tamaño de la Célula , Regulación hacia Abajo , Endorreduplicación , Conducta Alimentaria , Femenino , Células Germinativas/citología , Mitosis , Folículo Ovárico/citología , Folículo Ovárico/metabolismo , Células Madre/citología , Células Madre/metabolismo , Vitelogeninas/metabolismo
10.
Development ; 141(23): 4479-88, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25359724

RESUMEN

How adipocytes contribute to the physiological control of stem cells is a critical question towards understanding the link between obesity and multiple diseases, including cancers. Previous studies have revealed that adult stem cells are influenced by whole-body physiology through multiple diet-dependent factors. For example, nutrient-dependent pathways acting within the Drosophila ovary control the number and proliferation of germline stem cells (GSCs). The potential role of nutrient sensing by adipocytes in modulating stem cells in other organs, however, remains largely unexplored. Here, we report that amino acid sensing by adult adipocytes specifically modulates the maintenance of GSCs through a Target of Rapamycin-independent mechanism. Instead, reduced amino acid levels and the consequent increase in uncoupled tRNAs trigger activation of the GCN2-dependent amino acid response pathway within adipocytes, causing increased rates of GSC loss. These studies reveal a new step in adipocyte-stem cell crosstalk.


Asunto(s)
Adipocitos/fisiología , Células Madre Adultas/fisiología , Aminoácidos/metabolismo , Drosophila/fisiología , Transducción de Señal/fisiología , Adipocitos/metabolismo , Animales , Proteínas de Drosophila/metabolismo , Femenino , Microscopía Fluorescente , Ovario/citología , Proteínas Quinasas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina-Treonina Quinasas TOR/metabolismo
11.
Dev Biol ; 399(2): 226-36, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25576925

RESUMEN

Adipocytes have key endocrine roles, mediated in large part by secreted protein hormones termed adipokines. The adipokine adiponectin is well known for its role in sensitizing peripheral tissues to insulin, and several lines of evidence suggest that adiponectin might also modulate stem cells/precursors. It remains unclear, however, how adiponectin signaling controls stem cells and whether this role is secondary to its insulin-sensitizing effects or distinct. Drosophila adipocytes also function as an endocrine organ and, although no obvious adiponectin homolog has been identified, Drosophila AdipoR encodes a well-conserved homolog of mammalian adiponectin receptors. Here, we generate a null AdipoR allele and use clonal analysis to demonstrate an intrinsic requirement for AdipoR in germline stem cell (GSC) maintenance in the Drosophila ovary. AdipoR null GSCs are not fully responsive to bone morphogenetic protein ligands from the niche and have a slight reduction in E-cadherin levels at the GSC-niche junction. Conversely, germline-specific overexpression of AdipoR inhibits natural GSC loss, suggesting that reduction in adiponectin signaling might contribute to the normal decline in GSC numbers observed over time in wild-type females. Surprisingly, AdipoR is not required for insulin sensitization of the germline, leading us to speculate that insulin sensitization is a more recently acquired function than stem cell regulation in the evolutionary history of adiponectin signaling. Our findings establish Drosophila female GSCs as a new system for future studies addressing the molecular mechanisms whereby adiponectin receptor signaling modulates stem cell fate.


Asunto(s)
Adipocitos/fisiología , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Germinativas/citología , Receptores de Adiponectina/metabolismo , Transducción de Señal/fisiología , Células Madre/fisiología , Animales , Animales Modificados Genéticamente , Clonación Molecular , Cartilla de ADN/genética , Drosophila/citología , Proteínas de Drosophila/genética , Femenino , Procesamiento de Imagen Asistido por Computador , Insulina/metabolismo , Microscopía Fluorescente , Ovario/citología , Ovario/metabolismo , Receptores de Adiponectina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Dev Biol ; 400(1): 33-42, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25624267

RESUMEN

Nuclear hormone receptors have emerged as important regulators of mammalian and Drosophila adult physiology, affecting such seemingly diverse processes as adipogenesis, carbohydrate metabolism, circadian rhythm, stem cell function, and gamete production. Although nuclear hormone receptors Ecdysone Receptor (EcR) and Ultraspiracle (Usp) have multiple known roles in Drosophila development and regulate key processes during oogenesis, the adult function of the majority of nuclear hormone receptors remains largely undescribed. Ecdysone-induced protein 78C (E78), a nuclear hormone receptor closely related to Drosophila E75 and to mammalian Rev-Erb and Peroxisome Proliferator Activated Receptors, was originally identified as an early ecdysone target; however, it has remained unclear whether E78 significantly contributes to adult physiology or reproductive function. To further explore the biological function of E78 in oogenesis, we used available E78 reporters and created a new E78 loss-of-function allele. We found that E78 is expressed throughout the germline during oogenesis, and is important for proper egg production and for the maternal control of early embryogenesis. We showed that E78 is required during development to establish the somatic germline stem cell (GSC) niche, and that E78 function in the germline promotes the survival of developing follicles. Consistent with its initial discovery as an ecdysone-induced target, we also found significant genetic interactions between E78 and components of the ecdysone-signaling pathway. Taken together with the previously described roles of EcR, Usp, and E75, our results suggest that nuclear hormone receptors are critical for the broad transcriptional control of a wide variety of cellular processes during oogenesis.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/embriología , Células Madre Embrionarias/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Germinativas/fisiología , Oogénesis/fisiología , Folículo Ovárico/fisiología , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Cartilla de ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Galactósidos , Indoles , Microscopía Fluorescente , Interferencia de ARN , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo
13.
Development ; 140(3): 530-40, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23293285

RESUMEN

Stem cells must proliferate while maintaining 'stemness'; however, much remains to be learned about how factors that control the division of stem cells influence their identity. Multiple stem cell types display cell cycles with short G1 phases, thought to minimize susceptibility to differentiation factors. Drosophila female germline stem cells (GSCs) have short G1 and long G2 phases, and diet-dependent systemic factors often modulate G2. We previously observed that Cyclin E (CycE), a known G1/S regulator, is atypically expressed in GSCs during G2/M; however, it remained unclear whether CycE has cell cycle-independent roles in GSCs or whether it acts exclusively by modulating the cell cycle. In this study, we detected CycE activity during G2/M, reflecting its altered expression pattern, and showed that CycE and its canonical partner, Cyclin-dependent kinase 2 (Cdk2), are required not only for GSC proliferation, but also for GSC maintenance. In genetic mosaics, CycE- and Cdk2-deficient GSCs are rapidly lost from the niche, remain arrested in a G1-like state, and undergo excessive growth and incomplete differentiation. However, we found that CycE controls GSC maintenance independently of its role in the cell cycle; GSCs harboring specific hypomorphic CycE mutations are not efficiently maintained despite normal proliferation rates. Finally, CycE-deficient GSCs have an impaired response to niche bone morphogenetic protein signals that are required for GSC self-renewal, suggesting that CycE modulates niche-GSC communication. Taken together, these results show unequivocally that the roles of CycE/Cdk2 in GSC division cycle regulation and GSC maintenance are separable, and thus potentially involve distinct sets of phosphorylation targets.


Asunto(s)
Proliferación Celular , Ciclina E/metabolismo , Drosophila/citología , Nicho de Células Madre , Células Madre/citología , Alelos , Animales , Apoptosis , División Celular , Ciclina B/genética , Ciclina B/metabolismo , Ciclina E/genética , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Femenino , Fase G2 , Células Germinativas/citología , Células Germinativas/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Uniones Intercelulares/genética , Uniones Intercelulares/metabolismo , Mutación , Fenotipo , Transducción de Señal , Células Madre/metabolismo
14.
Development ; 137(13): 2117-26, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20504961

RESUMEN

Stem cells depend on intrinsic and local factors to maintain their identity and activity, but they also sense and respond to changing external conditions. We previously showed that germline stem cells (GSCs) and follicle stem cells (FSCs) in the Drosophila ovary respond to diet via insulin signals. Insulin signals directly modulate the GSC cell cycle at the G2 phase, but additional unknown dietary mediators control both G1 and G2. Target of rapamycin, or TOR, is part of a highly conserved nutrient-sensing pathway affecting growth, proliferation, survival and fertility. Here, we show that optimal TOR activity maintains GSCs but does not play a major role in FSC maintenance, suggesting differential regulation of GSCs versus FSCs. TOR promotes GSC proliferation via G2 but independently of insulin signaling, and TOR is required for the proliferation, growth and survival of differentiating germ cells. We also report that TOR controls the proliferation of FSCs but not of their differentiating progeny. Instead, TOR controls follicle cell number by promoting survival, independently of either the apoptotic or autophagic pathways. These results uncover specific TOR functions in the control of stem cells versus their differentiating progeny, and reveal parallels between Drosophila and mammalian follicle growth.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas Quinasas/metabolismo , Células Madre/metabolismo , Animales , Proliferación Celular , Drosophila melanogaster/metabolismo , Femenino , Insulina/metabolismo , Ovario/citología , Transducción de Señal , Serina-Treonina Quinasas TOR
15.
Sci Rep ; 13(1): 12331, 2023 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-37518578

RESUMEN

Temperature influences male fertility across organisms; however, how suboptimal temperatures affect adult spermatogenesis remains understudied. In a recent study on Drosophila melanogaster oogenesis, we observed a drastic reduction in the fertility of adult males exposed to warm temperature (29 °C). Here, we show that males become infertile at 29 °C because of low sperm abundance and quality. The low sperm abundance at 29 °C does not stem from reduced germline stem cell or spermatid numbers, as those numbers remain comparable between 29 °C and control 25 °C. Notably, males at cold 18 °C and 29 °C had similarly increased frequencies of spermatid elongation and individualization defects which, considering the high sperm abundance and male fertility measured at 18 °C, indicate that spermatogenesis has a high tolerance for elongation and individualization defects. Interestingly, the abundance of sperm at 29 °C decreases abruptly and with no evidence of apoptosis as they transition into the seminal vesicle near the end of spermatogenesis, pointing to sperm elimination through an unknown mechanism. Finally, sperm from males at 29 °C fertilize eggs less efficiently and do not support embryos past the first stage of embryogenesis, indicating that poor sperm quality is an additional cause of male infertility at 29 °C.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Animales , Masculino , Temperatura , Semen , Espermatozoides , Espermatogénesis , Fertilidad
16.
bioRxiv ; 2023 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-36712125

RESUMEN

Oligodendrocytes are the sole myelin producing cells in the central nervous system. Oligodendrocyte numbers are tightly controlled across diverse brain regions to match local axon type and number, but the underlying mechanisms and functional significance remain unclear. Here, we show that autophagy, an evolutionarily conserved cellular process that promotes cell survival under canonical settings, elicits premyelinating oligodendrocyte apoptosis during development and regulates critical aspects of nerve pulse propagation. Autophagy flux is increased in premyelinating oligodendrocytes, and its genetic blockage causes ectopic oligodendrocyte survival throughout the entire brain. Autophagy acts in the TFEB-Bax/Bak pathway and elevates PUMA mRNA levels to trigger premyelinating oligodendrocyte apoptosis cell-autonomously. Autophagy continuously functions in the myelinating oligodendrocytes to limit myelin sheath numbers and fine-tune nerve pulse propagation. Our results provide in vivo evidence showing that autophagy promotes apoptosis in mammalian cells under physiological conditions and reveal key intrinsic mechanisms governing oligodendrocyte number. HIGHLIGHTS: Autophagy flux increases in the premyelinating and myelinating oligodendrocytesAutophagy promotes premyelinating oligodendrocyte (pre-OL) apoptosis to control myelination location and timing Autophagy acts in the TFEB-PUMA-Bax/Bak pathway and elevates PUMA mRNA levels to determine pre-OL fate Autophagy continuously functions in the myelinating oligodendrocytes to limit myelin sheath thickness and finetune nerve pulse propagation.

17.
iScience ; 26(7): 107126, 2023 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-37426340

RESUMEN

TRIM24 is an oncogenic chromatin reader that is frequently overexpressed in human tumors and associated with poor prognosis. However, TRIM24 is rarely mutated, duplicated, or rearranged in cancer. This raises questions about how TRIM24 is regulated and what changes in its regulation are responsible for its overexpression. Here, we perform a genome-wide CRISPR-Cas9 screen by fluorescence-activated cell sorting (FACS) that nominated 220 negative regulators and elucidated a regulatory network that includes the KAP1 corepressor, CNOT deadenylase, and GID/CTLH E3 ligase. Knocking out required components of these three complexes caused TRIM24 overexpression, confirming their negative regulation of TRIM24. Our findings identify regulators of TRIM24 that nominate previously unexplored contexts for this oncoprotein in biology and disease. These findings were enabled by SLIDER, a new scoring system designed and vetted in our study as a broadly applicable tool for analysis of CRISPR screens performed by FACS.

18.
Cell Rep ; 42(8): 112943, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37543947

RESUMEN

Oligodendrocytes are the sole myelin-producing cells in the central nervous system. Oligodendrocyte number is tightly controlled across diverse brain regions to match local axon type and number, yet the underlying mechanisms remain unclear. Here, we show that autophagy, an evolutionarily conserved cellular process that promotes cell survival under physiological conditions, elicits premyelinating oligodendrocyte apoptosis during development. Autophagy flux is increased in premyelinating oligodendrocytes, and its genetic blockage causes ectopic oligodendrocyte survival throughout the entire brain. Autophagy functions cell autonomously in the premyelinating oligodendrocyte to trigger cell apoptosis, and it genetically interacts with the TFEB pathway to limit oligodendrocyte number across diverse brain regions. Our results provide in vivo evidence showing that autophagy promotes apoptosis in mammalian cells under physiological conditions and reveal key intrinsic mechanisms governing oligodendrogenesis.


Asunto(s)
Vaina de Mielina , Oligodendroglía , Animales , Oligodendroglía/metabolismo , Vaina de Mielina/metabolismo , Axones , Apoptosis , Autofagia , Diferenciación Celular/fisiología , Mamíferos
19.
Dev Biol ; 350(2): 290-300, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21145317

RESUMEN

Adult stem cells reside in specialized microenvironments, or niches, that are essential for their function in vivo. Stem cells are physically attached to the niche, which provides secreted factors that promote their self-renewal and proliferation. Despite intense research on the role of the niche in regulating stem cell function, much less is known about how the niche itself is controlled. We previously showed that insulin signals directly stimulate germline stem cell (GSC) division and indirectly promote GSC maintenance via the niche in Drosophila. Insulin-like peptides are required for maintenance of cap cells (a major component of the niche) via modulation of Notch signaling, and they also control attachment of GSCs to cap cells and E-cadherin levels at the cap cell-GSC junction. Here, we further dissect the molecular and cellular mechanisms underlying these processes. We show that insulin and Notch ligands directly stimulate cap cells to maintain their numbers and indirectly promote GSC maintenance. We also report that insulin signaling, via phosphoinositide 3-kinase and FOXO, intrinsically controls the competence of cap cells to respond to Notch ligands and thereby be maintained. Contrary to a previous report, we also find that Notch ligands originated in GSCs are not required either for Notch activation in the GSC niche, or for cap cell or GSC maintenance. Instead, the niche itself produces ligands that activate Notch signaling within cap cells, promoting stability of the GSC niche. Finally, insulin signals control cap cell-GSC attachment independently of their role in Notch signaling. These results are potentially relevant to many systems in which Notch signaling modulates stem cells and demonstrate that complex interactions between local and systemic signals are required for proper stem cell niche function.


Asunto(s)
Drosophila/embriología , Insulina/farmacología , Receptores Notch/fisiología , Transducción de Señal/fisiología , Nicho de Células Madre/fisiología , Animales , Proteínas de Drosophila/fisiología , Femenino , Factores de Transcripción Forkhead/fisiología , Ligandos , Fosfatidilinositol 3-Quinasas/fisiología , Receptor de Insulina/fisiología
20.
Proc Natl Acad Sci U S A ; 106(4): 1117-21, 2009 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-19136634

RESUMEN

Stem cell maintenance depends on local signals provided by specialized microenvironments, or niches, in which they reside. The potential role of systemic factors in stem cell maintenance, however, has remained largely unexplored. Here, we show that insulin signaling integrates the effects of diet and age on germline stem cell (GSC) maintenance through the dual regulation of cap cell number (via Notch signaling) and cap cell-GSC interaction (via E-cadherin) and that the normal process of GSC and niche cell loss that occurs with age can be suppressed by increased levels of insulin-like peptides. These results underscore the importance of systemic factors for the regulation of stem cell niches and, thereby, of stem cell numbers.


Asunto(s)
Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Insulina/metabolismo , Óvulo/citología , Nicho de Células Madre/citología , Nicho de Células Madre/metabolismo , Células Madre/citología , Animales , Cadherinas/metabolismo , Recuento de Células , Drosophila melanogaster/genética , Femenino , Genes de Insecto , Óvulo/metabolismo , Receptor de Insulina/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Células Madre/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA