Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Infect Immun ; 89(9): e0073420, 2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-33820816

RESUMEN

Along with respiratory tract disease per se, viral respiratory infections can also cause extrapulmonary complications with a potentially critical impact on health. In the present study, we used an experimental model of influenza A virus (IAV) infection to investigate the nature and outcome of the associated gut disorders. In IAV-infected mice, the signs of intestinal injury and inflammation, altered gene expression, and compromised intestinal barrier functions peaked on day 7 postinfection. As a likely result of bacterial component translocation, gene expression of inflammatory markers was upregulated in the liver. These changes occurred concomitantly with an alteration of the composition of the gut microbiota and with a decreased production of the fermentative, gut microbiota-derived products short-chain fatty acids (SCFAs). Gut inflammation and barrier dysfunction during influenza were not attributed to reduced food consumption, which caused in part gut dysbiosis. Treatment of IAV-infected mice with SCFAs was associated with an enhancement of intestinal barrier properties, as assessed by a reduction in the translocation of dextran and a decrease in inflammatory gene expression in the liver. Lastly, SCFA supplementation during influenza tended to reduce the translocation of the enteric pathogen Salmonella enterica serovar Typhimurium and to enhance the survival of doubly infected animals. Collectively, influenza virus infection can remotely impair the gut's barrier properties and trigger secondary enteric infections. The latter phenomenon can be partially countered by SCFA supplementation.


Asunto(s)
Infecciones por Enterobacteriaceae/etiología , Ácidos Grasos Volátiles/biosíntesis , Interacciones Huésped-Patógeno , Virus de la Influenza A/fisiología , Gripe Humana/complicaciones , Gripe Humana/virología , Mucosa Intestinal/metabolismo , Interacciones Microbianas , Susceptibilidad a Enfermedades , Disbiosis , Infecciones por Enterobacteriaceae/metabolismo , Interacciones Huésped-Patógeno/inmunología , Humanos , Gripe Humana/metabolismo , Mucosa Intestinal/inmunología
2.
Int J Mol Sci ; 23(1)2021 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-35008535

RESUMEN

Mitochondrial dysfunctions are implicated in several pathologies, such as metabolic, cardiovascular, respiratory, and neurological diseases, as well as in cancer and aging. These metabolic alterations are usually assessed in human or murine samples by mitochondrial respiratory chain enzymatic assays, by measuring the oxygen consumption of intact mitochondria isolated from tissues, or from cells obtained after physical or enzymatic disruption of the tissues. However, these methodologies do not maintain tissue multicellular organization and cell-cell interactions, known to influence mitochondrial metabolism. Here, we develop an optimal model to measure mitochondrial oxygen consumption in heart and lung tissue samples using the XF24 Extracellular Flux Analyzer (Seahorse) and discuss the advantages and limitations of this technological approach. Our results demonstrate that tissue organization, as well as mitochondrial ultrastructure and respiratory function, are preserved in heart and lung tissues freshly processed or after overnight conservation at 4 °C. Using this method, we confirmed the repeatedly reported obesity-associated mitochondrial dysfunction in the heart and extended it to the lungs. We set up and validated a new strategy to optimally assess mitochondrial function in murine tissues. As such, this method is of great potential interest for monitoring mitochondrial function in cohort samples.


Asunto(s)
Consumo de Oxígeno/fisiología , Envejecimiento/fisiología , Animales , Comunicación Celular/fisiología , Línea Celular , Línea Celular Tumoral , Respiración de la Célula/fisiología , Metabolismo Energético/fisiología , Corazón/fisiología , Humanos , Pulmón/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/fisiología , Membranas Mitocondriales/fisiología , Ratas , Pruebas de Función Respiratoria/métodos
3.
Nature ; 483(7389): 350-4, 2012 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-22343897

RESUMEN

Free fatty acids provide an important energy source as nutrients, and act as signalling molecules in various cellular processes. Several G-protein-coupled receptors have been identified as free-fatty-acid receptors important in physiology as well as in several diseases. GPR120 (also known as O3FAR1) functions as a receptor for unsaturated long-chain free fatty acids and has a critical role in various physiological homeostasis mechanisms such as adipogenesis, regulation of appetite and food preference. Here we show that GPR120-deficient mice fed a high-fat diet develop obesity, glucose intolerance and fatty liver with decreased adipocyte differentiation and lipogenesis and enhanced hepatic lipogenesis. Insulin resistance in such mice is associated with reduced insulin signalling and enhanced inflammation in adipose tissue. In human, we show that GPR120 expression in adipose tissue is significantly higher in obese individuals than in lean controls. GPR120 exon sequencing in obese subjects reveals a deleterious non-synonymous mutation (p.R270H) that inhibits GPR120 signalling activity. Furthermore, the p.R270H variant increases the risk of obesity in European populations. Overall, this study demonstrates that the lipid sensor GPR120 has a key role in sensing dietary fat and, therefore, in the control of energy balance in both humans and rodents.


Asunto(s)
Obesidad/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adipocitos/metabolismo , Adipocitos/patología , Adipogénesis , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Señalización del Calcio , Diferenciación Celular , Análisis Mutacional de ADN , Dieta Alta en Grasa , Metabolismo Energético , Europa (Continente)/etnología , Exones/genética , Hígado Graso/complicaciones , Hígado Graso/genética , Regulación de la Expresión Génica , Péptido 1 Similar al Glucagón/metabolismo , Glucosa/metabolismo , Intolerancia a la Glucosa/complicaciones , Humanos , Insulina/metabolismo , Resistencia a la Insulina , Lipogénesis , Hígado/metabolismo , Macrófagos/metabolismo , Ratones , Mutación/genética , Obesidad/complicaciones , Obesidad/genética , Obesidad/patología , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/genética , Población Blanca/genética
4.
Environ Microbiol ; 18(5): 1484-97, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26689997

RESUMEN

Alterations in gut microbiota composition and diversity were suggested to play a role in the development of obesity, a chronic subclinical inflammatory condition. We here evaluated the impact of oral consumption of a monostrain or multi-strain probiotic preparation in high-fat diet-induced obese mice. We observed a strain-specific effect and reported dissociation between the capacity of probiotics to dampen adipose tissue inflammation and to limit body weight gain. A multi-strain mixture was able to improve adiposity, insulin resistance and dyslipidemia through adipose tissue immune cell-remodelling, mainly affecting macrophages. At the gut level, the mixture modified the uptake of fatty acids and restored the expression level of the short-chain fatty acid receptor GPR43. These beneficial effects were associated with changes in the microbiota composition, such as the restoration of the abundance of Akkermansia muciniphila and Rikenellaceae and the decrease of other taxa like Lactobacillaceae. Using an in vitro gut model, we further showed that the probiotic mixture favours the production of butyrate and propionate. Our findings provide crucial clues for the design and use of more efficient probiotic preparations in obesity management and may bring new insights into the mechanisms by which host-microbe interactions govern such protective effects.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal/fisiología , Resistencia a la Insulina , Probióticos/uso terapéutico , Animales , Masculino , Ratones , Microbiota , Obesidad
5.
Stem Cells ; 31(4): 641-51, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23355370

RESUMEN

Melanoma is one of the most aggressive and extremely resistant to conventional therapies neoplasms. Recently, cellular resistance was linked to the cancer stem cell phenotype, still controversial and not well-defined. In this study, we used a Rhodamine 123 (Rh123) exclusion assay to functionally identify stem-like cells in metastatic human melanomas and melanoma cell lines. We demonstrate that a small subset of Rh123-low-retention (Rh123(low)) cells is enriched for stem cell-like activities, including the ability to self-renew and produce nonstem Rh123(high) progeny and to form melanospheres, recapitulating the phenotypic profile of the parental tumor. Rh123(low) cells are relatively quiescent and chemoresistant. At the molecular level, we show that melanoma Rh123(low) cells overexpress HIF1α, pluripotency factor OCT4, and the ABCB5 marker of melanoma stem cells and downregulate the expression of Cyclin D1 and CDK4. Interestingly, a short treatment with LY294002, an inhibitor of the PI3K/AKT pathway, specifically reverts a subset of Rh123(high) cells to the Rh123(low) phenotype, whereas treatment with inhibitors of mammalian target of rapamycin, phosphatase and tensin homolog or mitogen-activated protein kinase signaling does not. This phenotypic switching was associated with reduced levels of the HIF1α transcript and an increase in the level of phosphorylated nuclear FOXO3a preferentially in Rh123(low) cells. Moreover, the Rh123(low) cells became less quiescent and displayed a significant increase in their melanosphere-forming ability. All the above indicates that the Rh123(low) melanoma stem cell pool is composed of cycling and quiescent cells and that the PI3K/AKT signaling while maintaining the quiescence of Rh123(low) G0 cells promotes the exit of cycling cells from the stem cell compartment.


Asunto(s)
Melanoma/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Rodamina 123/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Cromonas/farmacología , Ciclina D1/genética , Ciclina D1/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunohistoquímica , Morfolinas/farmacología , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas
6.
Front Immunol ; 15: 1347676, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38590519

RESUMEN

The gut-lung axis is critical during viral respiratory infections such as influenza. Gut dysbiosis during infection translates into a massive drop of microbially produced short-chain fatty acids (SCFAs). Among them, butyrate is important during influenza suggesting that microbiome-based therapeutics targeting butyrate might hold promises. The butyrate-producing bacterium Faecalibacterium duncaniae (formerly referred to as F. prausnitzii) is an emerging probiotic with several health-promoting characteristics. To investigate the potential effects of F. duncaniae on influenza outcomes, mice were gavaged with live F. duncaniae (A2-165 or I-4574 strains) five days before infection. Supplementation of F. duncaniae was associated with less severe disease, a lower pulmonary viral load, and lower levels of lung inflammation. F. duncaniae supplementation impacted on gut dysbiosis induced by infection, as assessed by 16S rRNA sequencing. Interestingly, F. duncaniae administration was associated with a recovery in levels of SCFAs (including butyrate) in infected animals. The live form of F. duncaniae was more potent that the pasteurized form in improving influenza outcomes. Lastly, F. duncaniae partially protected against secondary (systemic) bacterial infection. We conclude that F. duncaniae might serve as a novel next generation probiotic against acute viral respiratory diseases.


Asunto(s)
Gripe Humana , Probióticos , Ratones , Animales , Humanos , Disbiosis/microbiología , ARN Ribosómico 16S/genética , Ácidos Grasos Volátiles , Butiratos , Faecalibacterium/genética
7.
Gut Microbes ; 16(1): 2325067, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38445660

RESUMEN

The gut-to-lung axis is critical during respiratory infections, including influenza A virus (IAV) infection. In the present study, we used high-resolution shotgun metagenomics and targeted metabolomic analysis to characterize influenza-associated changes in the composition and metabolism of the mouse gut microbiota. We observed several taxonomic-level changes on day (D)7 post-infection, including a marked reduction in the abundance of members of the Lactobacillaceae and Bifidobacteriaceae families, and an increase in the abundance of Akkermansia muciniphila. On D14, perturbation persisted in some species. Functional scale analysis of metagenomic data revealed transient changes in several metabolic pathways, particularly those leading to the production of short-chain fatty acids (SCFAs), polyamines, and tryptophan metabolites. Quantitative targeted metabolomics analysis of the serum revealed changes in specific classes of gut microbiota metabolites, including SCFAs, trimethylamine, polyamines, and indole-containing tryptophan metabolites. A marked decrease in indole-3-propionic acid (IPA) blood level was observed on D7. Changes in microbiota-associated metabolites correlated with changes in taxon abundance and disease marker levels. In particular, IPA was positively correlated with some Lactobacillaceae and Bifidobacteriaceae species (Limosilactobacillus reuteri, Lactobacillus animalis) and negatively correlated with Bacteroidales bacterium M7, viral load, and inflammation markers. IPA supplementation in diseased animals reduced viral load and lowered local (lung) and systemic inflammation. Treatment of mice with antibiotics targeting IPA-producing bacteria before infection enhanced viral load and lung inflammation, an effect inhibited by IPA supplementation. The results of this integrated metagenomic-metabolomic analysis highlighted IPA as an important contributor to influenza outcomes and a potential biomarker of disease severity.


Asunto(s)
Actinobacteria , Microbioma Gastrointestinal , Gripe Humana , Humanos , Animales , Ratones , Propionatos , Triptófano , Inflamación , Poliaminas
8.
Front Immunol ; 14: 1083191, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36936928

RESUMEN

Although the adipose tissue (AT) is a central metabolic organ in the regulation of whole-body energy homeostasis, it is also an important endocrine and immunological organ. As an endocrine organ, AT secretes a variety of bioactive peptides known as adipokines - some of which have inflammatory and immunoregulatory properties. As an immunological organ, AT contains a broad spectrum of innate and adaptive immune cells that have mostly been studied in the context of obesity. However, overwhelming evidence supports the notion that AT is a genuine immunological effector site, which contains all cell subsets required to induce and generate specific and effective immune responses against pathogens. Indeed, AT was reported to be an immune reservoir in the host's response to infection, and a site of parasitic, bacterial and viral infections. In addition, besides AT's immune cells, preadipocytes and adipocytes were shown to express innate immune receptors, and adipocytes were reported as antigen-presenting cells to regulate T-cell-mediated adaptive immunity. Here we review the current knowledge on the role of AT and AT's immune system in host defense against pathogens. First, we will summarize the main characteristics of AT: type, distribution, function, and extraordinary plasticity. Second, we will describe the intimate contact AT has with lymph nodes and vessels, and AT immune cell composition. Finally, we will present a comprehensive and up-to-date overview of the current research on the contribution of AT to host defense against pathogens, including the respiratory viruses influenza and SARS-CoV-2.


Asunto(s)
COVID-19 , Inmunidad Innata , Humanos , SARS-CoV-2 , Tejido Adiposo , Adipocitos/fisiología
9.
Cell Death Dis ; 14(2): 75, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36725844

RESUMEN

Coronavirus disease 2019 (COVID-19, caused by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2)) is primarily a respiratory illness. However, various extrapulmonary manifestations have been reported in patients with severe forms of COVID-19. Notably, SARS-CoV-2 was shown to directly trigger white adipose tissue (WAT) dysfunction, which in turn drives insulin resistance, dyslipidemia, and other adverse outcomes in patients with COVID-19. Although advanced age is the greatest risk factor for COVID-19 severity, published data on the impact of SARS-CoV-2 infection on WAT in aged individuals are scarce. Here, we characterized the response of subcutaneous and visceral WAT depots to SARS-CoV-2 infection in young adult and aged golden hamsters. In both age groups, infection was associated with a decrease in adipocyte size in the two WAT depots; this effect was partly due to changes in tissue's lipid metabolism and persisted for longer in aged hamsters than in young-adult hamsters. In contrast, only the subcutaneous WAT depot contained crown-like structures (CLSs) in which dead adipocytes were surrounded by SARS-CoV-2-infected macrophages, some of them forming syncytial multinucleated cells. Importantly, older age predisposed to a unique manifestation of viral disease in the subcutaneous WAT depot during SARS-CoV-2 infection; the persistence of very large CLSs was indicative of an age-associated defect in the clearance of dead adipocytes by macrophages. Moreover, we uncovered age-related differences in plasma lipid profiles during SARS-CoV-2 infection. These data suggest that the WAT's abnormal response to SARS-CoV-2 infection may contribute to the greater severity of COVID-19 observed in elderly patients.


Asunto(s)
Tejido Adiposo Blanco , COVID-19 , Animales , Cricetinae , Tejido Adiposo Blanco/patología , COVID-19/patología , Modelos Animales de Enfermedad , Mesocricetus , SARS-CoV-2
10.
Aging Cell ; 22(12): e14007, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37997569

RESUMEN

The age-related decline in immunity reduces the effectiveness of vaccines in older adults. Immunosenescence is associated with chronic, low-grade inflammation, and the accumulation of senescent cells. The latter express Bcl-2 family members (providing resistance to cell death) and exhibit a pro-inflammatory, senescence-associated secretory phenotype (SASP). Preexisting senescent cells cause many aging-related disorders and therapeutic means of eliminating these cells have recently gained attention. The potential consequences of senescent cell removal on vaccine efficacy in older individuals are still ignored. We used the Bcl-2 family inhibitor ABT-263 to investigate the effects of pre-vaccination senolysis on immune responses in old mice. Two different ovalbumin (OVA)-containing vaccines (containing a saponin-based or a CpG oligodeoxynucleotide adjuvant) were tested. ABT-263 depleted senescent cells (apoptosis) and ablated the basal and lipopolysaccharide-induced production of SASP-related factors in old mice. Depletion of senescent cells prior to vaccination (prime/boost) had little effect on OVA-specific antibody and T-cell responses (slightly reduced and augmented, respectively). We then used a preclinical melanoma model to test the antitumor potential of senolysis before vaccination (prime with the vaccine and OVA boost by tumor cells). Surprisingly, ABT-263 treatment abrogated the vaccine's ability to protect against B16 melanoma growth in old animals, an effect associated with reduced antigen-specific T-cell responses. Some, but not all, of the effects were age-specific, which suggests that preexisting senescent cells were partly involved. Hence, depletion of senescent cells modifies immune responses to vaccines in some settings and caution should be taken when incorporating senolytics into vaccine-based cancer therapies.


Asunto(s)
Vacunas contra el Cáncer , Vacunación , Animales , Ratones , Vacunas contra el Cáncer/farmacología , Senescencia Celular , Inmunidad , Proteínas Proto-Oncogénicas c-bcl-2
11.
Nat Aging ; 3(7): 829-845, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37414987

RESUMEN

Older age is one of the strongest risk factors for severe COVID-19. In this study, we determined whether age-associated cellular senescence contributes to the severity of experimental COVID-19. Aged golden hamsters accumulate senescent cells in the lungs, and the senolytic drug ABT-263, a BCL-2 inhibitor, depletes these cells at baseline and during SARS-CoV-2 infection. Relative to young hamsters, aged hamsters had a greater viral load during the acute phase of infection and displayed higher levels of sequelae during the post-acute phase. Early treatment with ABT-263 lowered pulmonary viral load in aged (but not young) animals, an effect associated with lower expression of ACE2, the receptor for SARS-CoV-2. ABT-263 treatment also led to lower pulmonary and systemic levels of senescence-associated secretory phenotype factors and to amelioration of early and late lung disease. These data demonstrate the causative role of age-associated pre-existing senescent cells on COVID-19 severity and have clear clinical relevance.


Asunto(s)
COVID-19 , SARS-CoV-2 , Cricetinae , Animales , Carga Viral , Pulmón , Mesocricetus , Inflamación , Senescencia Celular
12.
Am J Physiol Regul Integr Comp Physiol ; 303(2): R135-43, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22592557

RESUMEN

Human obesity is characterized by chronic low-grade inflammation in white adipose tissue and is often associated with hypertension. The potential induction of indoleamine 2,3-dioxygenase-1 (IDO1), the rate-limiting enzyme in tryptophan/kynurenine degradation pathway, by proinflammatory cytokines, could be associated with these disorders but has remained unexplored in obesity. Using immunohistochemistry, we detected IDO1 expression in white adipose tissue of obese patients, and we focused on its contribution in the regulation of vascular tone and on its immunoregulatory effects. Concentrations of tryptophan and kynurenine were measured in sera of 36 obese and 15 lean women. The expression of IDO1 in corresponding omental and subcutaneous adipose tissues and liver was evaluated. Proinflammatory markers and T-cell subsets were analyzed in adipose tissue via the expression of CD14, IL-18, CD68, TNFα, CD3ε, FOXP3 [a regulatory T-cell (Treg) marker] and RORC (a Th17 marker). In obese subjects, the ratio of kynurenine to tryptophan, which reflects IDO1 activation, is higher than in lean subjects. Furthermore, IDO1 expression in both adipose tissues and liver is increased and is inversely correlated with arterial blood pressure. Inflammation is associated with a T-cell infiltration in obese adipose tissue, with predominance of Th17 in the omental compartment and of Treg in the subcutaneous depot. The Th17/Treg balance is decreased in subcutaneous fat and correlates with IDO1 activation. In contrast, in the omental compartment, despite IDO1 activation, the Th17/Treg balance control is impaired. Taken together, our results suggest that IDO1 activation represents a local compensatory mechanism to limit obesity-induced inflammation and hypertension.


Asunto(s)
Tejido Adiposo/metabolismo , Presión Sanguínea/fisiología , Homeostasis/fisiología , Inmunidad/fisiología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Obesidad/metabolismo , Triptófano/sangre , Adulto , Estudios de Casos y Controles , Recuento de Células , Femenino , Humanos , Quinurenina/sangre , Hígado/metabolismo , Hígado/patología , Persona de Mediana Edad , Obesidad/patología , Obesidad/fisiopatología , Epiplón/metabolismo , Epiplón/patología , Transducción de Señal/fisiología , Linfocitos T Reguladores/patología , Células Th17/patología
13.
Viruses ; 14(9)2022 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-36146875

RESUMEN

Obese patients with non-alcoholic steatohepatitis (NASH) are prone to severe forms of COVID-19. There is an urgent need for new treatments that lower the severity of COVID-19 in this vulnerable population. To better replicate the human context, we set up a diet-induced model of obesity associated with dyslipidemia and NASH in the golden hamster (known to be a relevant preclinical model of COVID-19). A 20-week, free-choice diet induces obesity, dyslipidemia, and NASH (liver inflammation and fibrosis) in golden hamsters. Obese NASH hamsters have higher blood and pulmonary levels of inflammatory cytokines. In the early stages of a SARS-CoV-2 infection, the lung viral load and inflammation levels were similar in lean hamsters and obese NASH hamsters. However, obese NASH hamsters showed worse recovery (i.e., less resolution of lung inflammation 10 days post-infection (dpi) and lower body weight recovery on dpi 25). Obese NASH hamsters also exhibited higher levels of pulmonary fibrosis on dpi 25. Unlike lean animals, obese NASH hamsters infected with SARS-CoV-2 presented long-lasting dyslipidemia and systemic inflammation. Relative to lean controls, obese NASH hamsters had lower serum levels of angiotensin-converting enzyme 2 activity and higher serum levels of angiotensin II-a component known to favor inflammation and fibrosis. Even though the SARS-CoV-2 infection resulted in early weight loss and incomplete body weight recovery, obese NASH hamsters showed sustained liver steatosis, inflammation, hepatocyte ballooning, and marked liver fibrosis on dpi 25. We conclude that diet-induced obesity and NASH impair disease recovery in SARS-CoV-2-infected hamsters. This model might be of value for characterizing the pathophysiologic mechanisms of COVID-19 and evaluating the efficacy of treatments for the severe forms of COVID-19 observed in obese patients with NASH.


Asunto(s)
COVID-19 , Dislipidemias , Enfermedad del Hígado Graso no Alcohólico , Angiotensina II , Enzima Convertidora de Angiotensina 2 , Animales , COVID-19/complicaciones , Cricetinae , Citocinas , Dieta , Modelos Animales de Enfermedad , Humanos , Inflamación , Mesocricetus , Enfermedad del Hígado Graso no Alcohólico/etiología , Obesidad/complicaciones , SARS-CoV-2
14.
Gut Microbes ; 14(1): 2018900, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34965194

RESUMEN

Mounting evidence suggests that the gut-to-lung axis is critical during respiratory viral infections. We herein hypothesized that disruption of gut homeostasis during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection may associate with early disease outcomes. To address this question, we took advantage of the Syrian hamster model. Our data confirmed that this model recapitulates some hallmark features of the human disease in the lungs. We further showed that SARS-CoV-2 infection associated with mild intestinal inflammation, relative alteration in intestinal barrier property and liver inflammation and altered lipid metabolism. These changes occurred concomitantly with an alteration of the gut microbiota composition over the course of infection, notably characterized by a higher relative abundance of deleterious bacterial taxa such as Enterobacteriaceae and Desulfovibrionaceae. Conversely, several members of the Ruminococcaceae and Lachnospiraceae families, including bacteria known to produce the fermentative products short-chain fatty acids (SCFAs), had a reduced relative proportion compared to non-infected controls. Accordingly, infection led to a transient decrease in systemic SCFA amounts. SCFA supplementation during infection had no effect on clinical and inflammatory parameters. Lastly, a strong correlation between some gut microbiota taxa and clinical and inflammation indices of SARS-CoV-2 infection severity was evidenced. Collectively, alteration of the gut microbiota correlates with disease severity in hamsters making this experimental model valuable for the design of interventional, gut microbiota-targeted, approaches for the control of COVID-19.Abbreviations: SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; COVID-19, coronavirus disease 2019; SCFAs, short-chain fatty acids; dpi, day post-infection; RT-PCR, reverse transcription polymerase chain reaction; IL, interleukin. ACE2, angiotensin converting enzyme 2; TMPRSS2, transmembrane serine protease 2.


Asunto(s)
COVID-19/microbiología , COVID-19/fisiopatología , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Mesocricetus , Animales , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacterias/metabolismo , COVID-19/patología , Cricetinae , Ácidos Grasos Volátiles/administración & dosificación , Ácidos Grasos Volátiles/metabolismo , Humanos , Masculino , SARS-CoV-2/fisiología , Índice de Severidad de la Enfermedad , Tratamiento Farmacológico de COVID-19
16.
Nutrients ; 13(3)2021 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-33668212

RESUMEN

Since alterations of the gut microbiota have been shown to play a major role in obesity, probiotics have attracted attention. Our aim was to identify probiotic candidates for the management of obesity using a combination of in vitro and in vivo approaches. We evaluated in vitro the ability of 23 strains to limit lipid accumulation in adipocytes and to enhance the secretion of satiety-promoting gut peptide in enteroendocrine cells. Following the in vitro screening, selected strains were further investigated in vivo, single, or as mixtures, using a murine model of diet-induced obesity. Strain Bifidobacterium longum PI10 administrated alone and the mixture of B. animalis subsp. lactis LA804 and Lactobacillus gasseri LA806 limited body weight gain and reduced obesity-associated metabolic dysfunction and inflammation. These protective effects were associated with changes in the hypothalamic gene expression of leptin and leptin receptor as well as with changes in the composition of gut microbiota and the profile of bile acids. This study provides crucial clues to identify new potential probiotics as effective therapeutic approaches in the management of obesity, while also providing some insights into their mechanisms of action.


Asunto(s)
Adipocitos/microbiología , Células Enteroendocrinas/microbiología , Microbioma Gastrointestinal/fisiología , Obesidad/microbiología , Probióticos/farmacología , Animales , Ácidos y Sales Biliares/metabolismo , Dieta/efectos adversos , Modelos Animales de Enfermedad , Hormonas Gastrointestinales/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Ratones , Obesidad/etiología , Manejo de la Obesidad/métodos , Receptores de Leptina/metabolismo , Aumento de Peso/fisiología
17.
Differentiation ; 78(4): 223-31, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19682787

RESUMEN

Mesenchymal stem cells (MSC) are capable of both self-renewal and multi-lineage differentiation into mesoderm-type cells such as osteoblasts, chondrocytes, adipocytes and myocytes. Together the multipotent nature of MSCs and the facility to expand them in vitro make these cells ideal resources for regenerative medicine, particularly for bone reconstruction, and therefore research efforts focused on defining efficient protocols for directing their differentiation into the requisite lineage. Despite much progress in identifying mechanisms and factors that direct and control in vitro osteogenic differentiation of MSCs, a rapid and simple model to evaluate in vivo tissue formation is still lacking. Here, we describe the unique capacity of the murine bone marrow-derived D1 MSC cell line, which differentiates in vitro into at least three cell lineages, to form in vivo a structure resembling bone. This bone-like structure was obtained after subcutaneous grafting of D1 cells into immunocompetent mice without the need of neither an osteogenic factor nor scaffold material. These data allow us to propose this cell model as a tool for exploring in vivo the mechanisms and/or factors that govern and potentially regulate osteogenesis.


Asunto(s)
Diferenciación Celular , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Osteoblastos/citología , Osteogénesis , Animales , Linaje de la Célula , Células Cultivadas , Femenino , Histocitoquímica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones SCID , Osteoblastos/metabolismo , Organismos Libres de Patógenos Específicos
18.
Nutrients ; 12(3)2020 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-32244932

RESUMEN

Unhealthy lifestyle choices, such as bad eating behaviors and cigarette smoking, have major detrimental impacts on health. However, the inter-relations between obesity and smoking are still not fully understood. We thus developed an experimental model of high-fat diet-fed obese C57BL/6 male mice chronically exposed to cigarette smoke. Our study evaluated for the first time the resulting effects of the combined exposure to unhealthy diet and cigarette smoke on several metabolic, pulmonary, intestinal, and cardiac parameters. We showed that the chronic exposure to cigarette smoke modified the pattern of body fat distribution in favor of the visceral depots in obese mice, impaired the respiratory function, triggered pulmonary inflammation and emphysema, and was associated with gut microbiota dysbiosis, cardiac hypertrophy and myocardial fibrosis.


Asunto(s)
Exposición a Riesgos Ambientales , Estilo de Vida , Obesidad/etiología , Fumar/efectos adversos , Tejido Adiposo/metabolismo , Animales , Biomarcadores , Cardiomegalia/etiología , Cardiomegalia/metabolismo , Cardiomegalia/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Metabolismo Energético , Glucosa/metabolismo , Homeostasis , Humanos , Insulina/metabolismo , Pulmón/fisiopatología , Masculino , Ratones , Microbiota , Obesidad/complicaciones , Obesidad/metabolismo , Especificidad de Órganos
19.
Commun Biol ; 3(1): 237, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32409640

RESUMEN

Like all obligate intracellular pathogens, influenza A virus (IAV) reprograms host cell's glucose and lipid metabolism to promote its own replication. However, the impact of influenza infection on white adipose tissue (WAT), a key tissue in the control of systemic energy homeostasis, has not been yet characterized. Here, we show that influenza infection induces alterations in whole-body glucose metabolism that persist long after the virus has been cleared. We report depot-specific changes in the WAT of IAV-infected mice, notably characterized by the appearance of thermogenic brown-like adipocytes within the subcutaneous fat depot. Importantly, viral RNA- and viral antigen-harboring cells are detected in the WAT of infected mice. Using in vitro approaches, we find that IAV infection enhances the expression of brown-adipogenesis-related genes in preadipocytes. Overall, our findings shed light on the role that the white adipose tissue, which lies at the crossroads of nutrition, metabolism and immunity, may play in influenza infection.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Metabolismo Energético , Infecciones por Orthomyxoviridae/metabolismo , Termogénesis , Tejido Adiposo Pardo/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Gripe Humana/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
20.
Cell Rep ; 30(9): 2934-2947.e6, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32130898

RESUMEN

Secondary bacterial infections often complicate viral respiratory infections. We hypothesize that perturbation of the gut microbiota during influenza A virus (IAV) infection might favor respiratory bacterial superinfection. Sublethal infection with influenza transiently alters the composition and fermentative activity of the gut microbiota in mice. These changes are attributed in part to reduced food consumption. Fecal transfer experiments demonstrate that the IAV-conditioned microbiota compromises lung defenses against pneumococcal infection. In mechanistic terms, reduced production of the predominant short-chain fatty acid (SCFA) acetate affects the bactericidal activity of alveolar macrophages. Following treatment with acetate, mice colonized with the IAV-conditioned microbiota display reduced bacterial loads. In the context of influenza infection, acetate supplementation reduces, in a free fatty acid receptor 2 (FFAR2)-dependent manner, local and systemic bacterial loads. This translates into reduced lung pathology and improved survival rates of double-infected mice. Lastly, pharmacological activation of the SCFA receptor FFAR2 during influenza reduces bacterial superinfection.


Asunto(s)
Disbiosis/microbiología , Ácidos Grasos Volátiles/biosíntesis , Tracto Gastrointestinal/microbiología , Gripe Humana/microbiología , Pulmón/microbiología , Infecciones Neumocócicas/complicaciones , Sobreinfección/complicaciones , Sobreinfección/microbiología , Acetatos/farmacología , Animales , Disbiosis/complicaciones , Disbiosis/virología , Conducta Alimentaria , Microbioma Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/efectos de los fármacos , Humanos , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/microbiología , Macrófagos Alveolares/patología , Ratones Endogámicos C57BL , Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/virología , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Infecciones del Sistema Respiratorio/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA