14 research outputs found

    Reproductive capacity of the red cusk-eel genypterus chilensis (Guichenot, 1848) in captivity

    Get PDF
    Indexación: Scopus.This work was supported by the FONDEF Project D06I 1024 “Development of technologies for the production of red cusk-eel fingerlings (Genypterus chilensis)”.Genypterus chilensis is a marine fish of high gastronomic demand, whose capture has declined in recent years due to overfishing. In the development of the farming technology, high mortalities were obtained during egg incubation. The objective of this study is to contribute to the knowledge of fecundity and eggs viability of G. chilensis in captivity. The spawns of G. chilensis were analyzed over a period of 2 years and 3 months. The total fecundity was estimated by counting the masses and eggs produced monthly throughout the period. The results confirm that G. chilensis is a partial spawner, since a female may more than two masses of eggs per day, due to a large amount of mass spawned per season (621 average). The total production of masses of the Farming Centre during the period was 2,290; of these, only 7% (166) corresponding to 15,330,517 eggs were incubated. Because of its high fecundity, G. chilensis produces numerous masses of eggs, of which only a small percentage reaches incubation, as well as it occurs in other marine fish. © 2018, Escuela de Ciencias del Mar. All rights reserved.https://scielo.conicyt.cl/scielo.php?script=sci_arttext&pid=S0718-560X201800020048

    50 Years of Cetacean Strandings Reveal a Concerning Rise in Chilean Patagonia

    Get PDF
    Indexación ScopusCetacean strandings (CS) have been reported in increasing numbers in coastal areas worldwide. Although the causes of these strandings are unknown, a number of anthropogenic and environmental factors have been suggested. This paper aims to characterize CS patterns and describe their fine-scale spatiotemporal dynamics. We analysed spatial and spatiotemporal CS patterns in Chile from January 1968 to January 2020. We identified a total of 389 CS events affecting eight cetacean families, 21 genera, and 35 species, which represent more than 85% of the reported species richness for the country. Most CS events (94.1%) were single (i.e., ≤two individuals). There were also 18 mass stranding (three to 24 individuals, 4.1%) and nine unusually large mass stranding events (>25 individuals, 2%). Purely spatial tests showed CS events appearing in random occurrence along the Chilean coast. Local tests for spatio-temporal clusters, however, identified a greater number of hotspots reported in the southernmost part of the country, namely, Chilean Patagonia. Specifically, significant spatio-temporal clusters were identified and defined as containing three or more individuals within a two-month period as a focal coastal event (<1 km radius). It is a cause of concern that CS events in Chile have been increasing consistently over the last decades, and although we were not able to identify their causes, we are able to highlight the importance of changes in climate conditions and of an increase in monitoring activities as primary drivers for such patterns, particularly important in Chilean Patagonia. © 2020, The Author(s).https://www-nature-com.recursosbiblioteca.unab.cl/articles/s41598-020-66484-

    Consensus on complementary feeding from the Latin American Society for Pediatric Gastroenterology, Hepatology and Nutrition: COCO 2023

    Get PDF
    Abstract Complementary feeding (CF) is defined as infant feeding that complements breastfeeding or, alternatively, breastfeeding with a breast milk substitute, and is a process that goes beyond simply providing guidance on what and how to introduce foods. The information provided by health professionals should be up-to-date and evidence-based. There are different guidelines or position papers at the international level, which, although most of the recommendations may be applicable, there are some others that require regionalization or adaptation to the conditions and reality of each area. The Nutrition working group of the Latin American Society of Pediatric Gastroenterology, Hepatology and Nutrition convened a group of experts, representatives from each of the countries that make up the society, with the objective of developing a consensus on CA, incorporating, when possible, local information that adapts to the reality of the region. The purpose of this document is to show the results of this work. Through Delphi methodology, a total of 34 statements or statements regarding relevant aspects of CA were evaluated, discussed and voted upon.Resumen La alimentación complementaria (AC) se define como la alimentación de los lactantes que complementa a la lactancia materna o en su defecto, a la lactancia con un sucedáneo de la leche materna, y es un proceso que va más allá de simplemente una guía sobre qué y cómo introducir los alimentos. La información brindada por parte de los profesionales de la salud debe ser actualizada y basada en evidencia. Existen diferentes guías o documentos de posición a nivel internacional, que, aunque la mayoría de las recomendaciones pueden ser aplicables, hay algunas otras que requieren una regionalización o adecuación a las condiciones y realidad de cada zona. El grupo de trabajo de Nutrición de la Sociedad Latinoamericana de Gastroenterología, Hepatología y Nutrición Pediátrica convocó a un grupo de expertos, representantes de cada uno de los países que conforman la sociedad, con el objetivo de desarrollar un consenso sobre la AC, que incorporó cuando así fue posible, información local que se adapte a la realidad de la región. El objetivo de este documento es mostrar los resultados de dicho trabajo. A través de metodología Delphi, se evaluaron, discutieron y votaron un total de 34 declaraciones o enunciados con respecto a aspectos relevantes de la AC

    Image_6_Human metapneumovirus respiratory infection affects both innate and adaptive intestinal immunity.jpeg

    No full text
    IntroductionRespiratory infections are one of the leading causes of morbidity and mortality worldwide, mainly in children, immunocompromised people, and the elderly. Several respiratory viruses can induce intestinal inflammation and alterations in intestinal microbiota composition. Human metapneumovirus (HMPV) is one of the major respiratory viruses contributing to infant mortality in children under 5 years of age worldwide, and the effect of this infection at the gut level has not been studied.MethodsHere, we evaluated the distal effects of HMPV infection on intestinal microbiota and inflammation in a murine model, analyzing several post-infection times (days 1, 3, and 5). Six to eight-week-old C57BL/6 mice were infected intranasally with HMPV, and mice inoculated with a non-infectious supernatant (Mock) were used as a control group.ResultsWe did not detect HMPV viral load in the intestine, but we observed significant changes in the transcription of IFN-γ in the colon, analyzed by qPCR, at day 1 post-infection as compared to the control group. Furthermore, we analyzed the frequencies of different innate and adaptive immune cells in the colonic lamina propria, using flow cytometry. The frequency of monocyte populations was altered in the colon of HMPV -infected mice at days 1 and 3, with no significant difference from control mice at day 5 post-infection. Moreover, colonic CD8+ T cells and memory precursor effector CD8+ T cells were significantly increased in HMPV-infected mice at day 5, suggesting that HMPV may also alter intestinal adaptive immunity. Additionally, we did not find alterations in antimicrobial peptide expression, the frequency of colonic IgA+ plasma cells, and levels of fecal IgA. Some minor alterations in the fecal microbiota composition of HMPV -infected mice were detected using 16s rRNA sequencing. However, no significant differences were found in β-diversity and relative abundance at the genus level.DiscussionTo our knowledge, this is the first report describing the alterations in intestinal immunity following respiratory infection with HMPV infection. These effects do not seem to be mediated by direct viral infection in the intestinal tract. Our results indicate that HMPV can affect colonic innate and adaptive immunity but does not significantly alter the microbiota composition, and further research is required to understand the mechanisms inducing these distal effects in the intestine.</p

    Image_3_Human metapneumovirus respiratory infection affects both innate and adaptive intestinal immunity.jpeg

    No full text
    IntroductionRespiratory infections are one of the leading causes of morbidity and mortality worldwide, mainly in children, immunocompromised people, and the elderly. Several respiratory viruses can induce intestinal inflammation and alterations in intestinal microbiota composition. Human metapneumovirus (HMPV) is one of the major respiratory viruses contributing to infant mortality in children under 5 years of age worldwide, and the effect of this infection at the gut level has not been studied.MethodsHere, we evaluated the distal effects of HMPV infection on intestinal microbiota and inflammation in a murine model, analyzing several post-infection times (days 1, 3, and 5). Six to eight-week-old C57BL/6 mice were infected intranasally with HMPV, and mice inoculated with a non-infectious supernatant (Mock) were used as a control group.ResultsWe did not detect HMPV viral load in the intestine, but we observed significant changes in the transcription of IFN-γ in the colon, analyzed by qPCR, at day 1 post-infection as compared to the control group. Furthermore, we analyzed the frequencies of different innate and adaptive immune cells in the colonic lamina propria, using flow cytometry. The frequency of monocyte populations was altered in the colon of HMPV -infected mice at days 1 and 3, with no significant difference from control mice at day 5 post-infection. Moreover, colonic CD8+ T cells and memory precursor effector CD8+ T cells were significantly increased in HMPV-infected mice at day 5, suggesting that HMPV may also alter intestinal adaptive immunity. Additionally, we did not find alterations in antimicrobial peptide expression, the frequency of colonic IgA+ plasma cells, and levels of fecal IgA. Some minor alterations in the fecal microbiota composition of HMPV -infected mice were detected using 16s rRNA sequencing. However, no significant differences were found in β-diversity and relative abundance at the genus level.DiscussionTo our knowledge, this is the first report describing the alterations in intestinal immunity following respiratory infection with HMPV infection. These effects do not seem to be mediated by direct viral infection in the intestinal tract. Our results indicate that HMPV can affect colonic innate and adaptive immunity but does not significantly alter the microbiota composition, and further research is required to understand the mechanisms inducing these distal effects in the intestine.</p

    Image_4_Human metapneumovirus respiratory infection affects both innate and adaptive intestinal immunity.jpeg

    No full text
    IntroductionRespiratory infections are one of the leading causes of morbidity and mortality worldwide, mainly in children, immunocompromised people, and the elderly. Several respiratory viruses can induce intestinal inflammation and alterations in intestinal microbiota composition. Human metapneumovirus (HMPV) is one of the major respiratory viruses contributing to infant mortality in children under 5 years of age worldwide, and the effect of this infection at the gut level has not been studied.MethodsHere, we evaluated the distal effects of HMPV infection on intestinal microbiota and inflammation in a murine model, analyzing several post-infection times (days 1, 3, and 5). Six to eight-week-old C57BL/6 mice were infected intranasally with HMPV, and mice inoculated with a non-infectious supernatant (Mock) were used as a control group.ResultsWe did not detect HMPV viral load in the intestine, but we observed significant changes in the transcription of IFN-γ in the colon, analyzed by qPCR, at day 1 post-infection as compared to the control group. Furthermore, we analyzed the frequencies of different innate and adaptive immune cells in the colonic lamina propria, using flow cytometry. The frequency of monocyte populations was altered in the colon of HMPV -infected mice at days 1 and 3, with no significant difference from control mice at day 5 post-infection. Moreover, colonic CD8+ T cells and memory precursor effector CD8+ T cells were significantly increased in HMPV-infected mice at day 5, suggesting that HMPV may also alter intestinal adaptive immunity. Additionally, we did not find alterations in antimicrobial peptide expression, the frequency of colonic IgA+ plasma cells, and levels of fecal IgA. Some minor alterations in the fecal microbiota composition of HMPV -infected mice were detected using 16s rRNA sequencing. However, no significant differences were found in β-diversity and relative abundance at the genus level.DiscussionTo our knowledge, this is the first report describing the alterations in intestinal immunity following respiratory infection with HMPV infection. These effects do not seem to be mediated by direct viral infection in the intestinal tract. Our results indicate that HMPV can affect colonic innate and adaptive immunity but does not significantly alter the microbiota composition, and further research is required to understand the mechanisms inducing these distal effects in the intestine.</p

    Image_1_Human metapneumovirus respiratory infection affects both innate and adaptive intestinal immunity.jpeg

    No full text
    IntroductionRespiratory infections are one of the leading causes of morbidity and mortality worldwide, mainly in children, immunocompromised people, and the elderly. Several respiratory viruses can induce intestinal inflammation and alterations in intestinal microbiota composition. Human metapneumovirus (HMPV) is one of the major respiratory viruses contributing to infant mortality in children under 5 years of age worldwide, and the effect of this infection at the gut level has not been studied.MethodsHere, we evaluated the distal effects of HMPV infection on intestinal microbiota and inflammation in a murine model, analyzing several post-infection times (days 1, 3, and 5). Six to eight-week-old C57BL/6 mice were infected intranasally with HMPV, and mice inoculated with a non-infectious supernatant (Mock) were used as a control group.ResultsWe did not detect HMPV viral load in the intestine, but we observed significant changes in the transcription of IFN-γ in the colon, analyzed by qPCR, at day 1 post-infection as compared to the control group. Furthermore, we analyzed the frequencies of different innate and adaptive immune cells in the colonic lamina propria, using flow cytometry. The frequency of monocyte populations was altered in the colon of HMPV -infected mice at days 1 and 3, with no significant difference from control mice at day 5 post-infection. Moreover, colonic CD8+ T cells and memory precursor effector CD8+ T cells were significantly increased in HMPV-infected mice at day 5, suggesting that HMPV may also alter intestinal adaptive immunity. Additionally, we did not find alterations in antimicrobial peptide expression, the frequency of colonic IgA+ plasma cells, and levels of fecal IgA. Some minor alterations in the fecal microbiota composition of HMPV -infected mice were detected using 16s rRNA sequencing. However, no significant differences were found in β-diversity and relative abundance at the genus level.DiscussionTo our knowledge, this is the first report describing the alterations in intestinal immunity following respiratory infection with HMPV infection. These effects do not seem to be mediated by direct viral infection in the intestinal tract. Our results indicate that HMPV can affect colonic innate and adaptive immunity but does not significantly alter the microbiota composition, and further research is required to understand the mechanisms inducing these distal effects in the intestine.</p

    Image_5_Human metapneumovirus respiratory infection affects both innate and adaptive intestinal immunity.jpeg

    No full text
    IntroductionRespiratory infections are one of the leading causes of morbidity and mortality worldwide, mainly in children, immunocompromised people, and the elderly. Several respiratory viruses can induce intestinal inflammation and alterations in intestinal microbiota composition. Human metapneumovirus (HMPV) is one of the major respiratory viruses contributing to infant mortality in children under 5 years of age worldwide, and the effect of this infection at the gut level has not been studied.MethodsHere, we evaluated the distal effects of HMPV infection on intestinal microbiota and inflammation in a murine model, analyzing several post-infection times (days 1, 3, and 5). Six to eight-week-old C57BL/6 mice were infected intranasally with HMPV, and mice inoculated with a non-infectious supernatant (Mock) were used as a control group.ResultsWe did not detect HMPV viral load in the intestine, but we observed significant changes in the transcription of IFN-γ in the colon, analyzed by qPCR, at day 1 post-infection as compared to the control group. Furthermore, we analyzed the frequencies of different innate and adaptive immune cells in the colonic lamina propria, using flow cytometry. The frequency of monocyte populations was altered in the colon of HMPV -infected mice at days 1 and 3, with no significant difference from control mice at day 5 post-infection. Moreover, colonic CD8+ T cells and memory precursor effector CD8+ T cells were significantly increased in HMPV-infected mice at day 5, suggesting that HMPV may also alter intestinal adaptive immunity. Additionally, we did not find alterations in antimicrobial peptide expression, the frequency of colonic IgA+ plasma cells, and levels of fecal IgA. Some minor alterations in the fecal microbiota composition of HMPV -infected mice were detected using 16s rRNA sequencing. However, no significant differences were found in β-diversity and relative abundance at the genus level.DiscussionTo our knowledge, this is the first report describing the alterations in intestinal immunity following respiratory infection with HMPV infection. These effects do not seem to be mediated by direct viral infection in the intestinal tract. Our results indicate that HMPV can affect colonic innate and adaptive immunity but does not significantly alter the microbiota composition, and further research is required to understand the mechanisms inducing these distal effects in the intestine.</p

    Image_2_Human metapneumovirus respiratory infection affects both innate and adaptive intestinal immunity.jpeg

    No full text
    IntroductionRespiratory infections are one of the leading causes of morbidity and mortality worldwide, mainly in children, immunocompromised people, and the elderly. Several respiratory viruses can induce intestinal inflammation and alterations in intestinal microbiota composition. Human metapneumovirus (HMPV) is one of the major respiratory viruses contributing to infant mortality in children under 5 years of age worldwide, and the effect of this infection at the gut level has not been studied.MethodsHere, we evaluated the distal effects of HMPV infection on intestinal microbiota and inflammation in a murine model, analyzing several post-infection times (days 1, 3, and 5). Six to eight-week-old C57BL/6 mice were infected intranasally with HMPV, and mice inoculated with a non-infectious supernatant (Mock) were used as a control group.ResultsWe did not detect HMPV viral load in the intestine, but we observed significant changes in the transcription of IFN-γ in the colon, analyzed by qPCR, at day 1 post-infection as compared to the control group. Furthermore, we analyzed the frequencies of different innate and adaptive immune cells in the colonic lamina propria, using flow cytometry. The frequency of monocyte populations was altered in the colon of HMPV -infected mice at days 1 and 3, with no significant difference from control mice at day 5 post-infection. Moreover, colonic CD8+ T cells and memory precursor effector CD8+ T cells were significantly increased in HMPV-infected mice at day 5, suggesting that HMPV may also alter intestinal adaptive immunity. Additionally, we did not find alterations in antimicrobial peptide expression, the frequency of colonic IgA+ plasma cells, and levels of fecal IgA. Some minor alterations in the fecal microbiota composition of HMPV -infected mice were detected using 16s rRNA sequencing. However, no significant differences were found in β-diversity and relative abundance at the genus level.DiscussionTo our knowledge, this is the first report describing the alterations in intestinal immunity following respiratory infection with HMPV infection. These effects do not seem to be mediated by direct viral infection in the intestinal tract. Our results indicate that HMPV can affect colonic innate and adaptive immunity but does not significantly alter the microbiota composition, and further research is required to understand the mechanisms inducing these distal effects in the intestine.</p
    corecore