Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
2.
Hepatology ; 77(5): 1654-1669, 2023 05 01.
Article in English | MEDLINE | ID: mdl-35921199

ABSTRACT

BACKGROUND AND AIMS: Recent studies suggest that mitochondrial dysfunction promotes progression to NASH by aggravating the gut-liver status. However, the underlying mechanism remains unclear. Herein, we hypothesized that enhanced mitochondrial activity might reshape a specific microbiota signature that, when transferred to germ-free (GF) mice, could delay NASH progression. APPROACH AND RESULTS: Wild-type and methylation-controlled J protein knockout (MCJ-KO) mice were fed for 6 weeks with either control or a choline-deficient, L-amino acid-defined, high-fat diet (CDA-HFD). One mouse of each group acted as a donor of cecal microbiota to GF mice, who also underwent the CDA-HFD model for 3 weeks. Hepatic injury, intestinal barrier, gut microbiome, and the associated fecal metabolome were then studied. Following 6 weeks of CDA-HFD, the absence of methylation-controlled J protein, an inhibitor of mitochondrial complex I activity, reduced hepatic injury and improved gut-liver axis in an aggressive NASH dietary model. This effect was transferred to GF mice through cecal microbiota transplantation. We suggest that the specific microbiota profile of MCJ-KO, characterized by an increase in the fecal relative abundance of Dorea and Oscillospira genera and a reduction in AF12 , Allboaculum , and [ Ruminococcus ], exerted protective actions through enhancing short-chain fatty acids, nicotinamide adenine dinucleotide (NAD + ) metabolism, and sirtuin activity, subsequently increasing fatty acid oxidation in GF mice. Importantly, we identified Dorea genus as one of the main modulators of this microbiota-dependent protective phenotype. CONCLUSIONS: Overall, we provide evidence for the relevance of mitochondria-microbiota interplay during NASH and that targeting it could be a valuable therapeutic approach.


Subject(s)
Gastrointestinal Microbiome , Non-alcoholic Fatty Liver Disease , Mice , Animals , Non-alcoholic Fatty Liver Disease/metabolism , Gastrointestinal Microbiome/genetics , Mice, Inbred C57BL , Liver/metabolism , Diet, High-Fat/adverse effects , Molecular Chaperones/metabolism , Mitochondrial Proteins/metabolism
3.
Medicine (Baltimore) ; 101(33): e29954, 2022 Aug 19.
Article in English | MEDLINE | ID: mdl-35984207

ABSTRACT

This observational, cross-sectional case-control study evaluates the impact of coronavirus disease 2019 (COVID-19) on health-related quality of life (HRQoL) in elderly persons who have undergone surgery for adult spinal deformity (ASD). On December 31, 2019, the Chinese authorities first reported severe acute respiratory syndrome coronavirus 2, and on March 11, 2020, it was declared a pandemic. The pandemic seems to have had a negative effect on elderly patients who underwent ASD, in terms of functional and psychological quality of life. We selected patients with ASD aged > 70 years who had undergone surgery between 2010 and 2015 and compared them with age- and sex-matched patients who did not have ASD. We recorded sociodemographic variables, type of surgery, levels of spinal fusion, HRQoL (Scoliosis Research Society-22, Short Form 12 Health Survey, EuroQol-5D [EQ-5], Geriatric Depression Scale [Yesavage] [GDS], Modified Frailty Index-11, and Barthel index), fear of visiting a health center, fear of leaving one's house, and adherence to preventive measures. The study population comprised 174 patients (mean [standard deviation] age, 77.3 [5.9] years; 86% women), of whom 87 had undergone surgery for ASD. The incidence of COVID-19 was higher in patients aged > 85 years (P = .041), urban areas (P = .047), and in patients in long-term care (P = .03). Similarly, no differences were observed for the ability to cope with the pandemic (P > .05). Patients who underwent surgery also had a higher risk of depression (GDS, 6.7 [P = .02]), a lower EQ-5 score (P = .001), a higher body mass index (P = .004), greater consumption of drugs (P < .001), especially opiates (P < .001). Patients who underwent surgery constitute a vulnerable population during the COVID-19 pandemic, with poorer quality of life and had a much higher risk of depression. They are also polymedicated and prefrail, adhere well to COVID-19 preventive measures, and do not seem to fear visiting health centers.


Subject(s)
COVID-19 , Quality of Life , Adult , Aged , COVID-19/epidemiology , Case-Control Studies , Cross-Sectional Studies , Female , Follow-Up Studies , Humans , Male , Pandemics , Retrospective Studies
4.
Antioxidants (Basel) ; 10(12)2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34943104

ABSTRACT

Gut microbiota plays a key role in obesity and non-alcoholic fatty liver disease (NAFLD), so synbiotics could be a therapeutic alternative. We aim to evaluate a nutritional intervention together with the administration of the bacteria Akkermansia muciniphila and the antioxidant quercetin in an in vivo model of early obesity and NAFLD. 21-day-old rats were fed with control or high-fat diet for six weeks. Then, all animals received control diet supplemented with/without quercetin and/or A. muciniphila for three weeks. Gut microbiota, NAFLD-related parameters, circulating bile acids (BAs) and liver gene expression were analyzed. The colonization with A. muciniphila was associated with less body fat, while synbiotic treatment caused a steatosis remission, linked to hepatic lipogenesis modulation. The synbiotic promoted higher abundance of Cyanobacteria and Oscillospira, and lower levels of Actinobacteria, Lactococcus, Lactobacillus and Roseburia. Moreover, it favored elevated unconjugated hydrophilic BAs plasma levels and enhanced hepatic expression of BA synthesis and transport genes. A. muciniphila correlated with circulating BAs and liver lipid and BA metabolism genes, suggesting a role of this bacterium in BA signaling. Beneficial effects of A. muciniphila and quercetin combination are driven by gut microbiota modulation, the shift in BAs and the gut-liver bile flow enhancement.

5.
Food Chem Toxicol ; 158: 112664, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34767876

ABSTRACT

Treatment of ß-lactamase positive bacterial infections with a combination of amoxicillin (AMOX) and clavulanic acid (CLAV) causes idiosyncratic drug-induced liver injury (iDILI) in a relevant number of patients, often with features of intrahepatic cholestasis. This study aims to determine serum bile acid (BA) levels in amoxicillin/clavulanate (A+C)-iDILI patients and to investigate the mechanism of cholestasis by A+C in human in vitro hepatic models. In six A+C-iDILI patients, significant elevations of serum primary conjugated BA definitely demonstrated A+C-induced cholestasis. In cultured human Upcyte hepatocytes and HepG2 cells, CLAV was more cytotoxic than AMOX, and, at subcytotoxic concentrations, it altered the expression of more than 1,300 genes. CLAV, but not AMOX, downregulated the expression of key genes for BA transport (BSEP, NTCP, OSTα and MDR2) and synthesis (CYP7A1 and CYP8B1). CLAV also caused early oxidative stress, with reduced GSH/GSSG ratio, along with induction of antioxidant nuclear factor erythroid 2-related factor 2 (NRF2) target genes. Activation of NRF2 by sulforaphane also resulted in downregulation of NTCP, OSTα, ABCG5, CYP7A1 and CYP8B1. CLAV also inhibited the BA-sensor farnesoid X receptor (FXR), in agreement with the downregulation of FXR targets BSEP, OSTα and ABCG5. We conclude that CLAV, the culprit molecule in A+C, downregulates several key biliary transporters by modulating NRF2 and FXR signaling, thus likely promoting intrahepatic cholestasis. On top of that, increased ROS production and GSH depletion may aggravate the cholestatic injury by A+C.


Subject(s)
Cholestasis, Intrahepatic , Clavulanic Acid/toxicity , NF-E2-Related Factor 2 , Receptors, Cytoplasmic and Nuclear , Aged , Cell Line , Cholestasis, Intrahepatic/chemically induced , Cholestasis, Intrahepatic/genetics , Cholestasis, Intrahepatic/metabolism , Female , Humans , Male , Middle Aged , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
6.
Nutrients ; 13(8)2021 Jul 23.
Article in English | MEDLINE | ID: mdl-34444679

ABSTRACT

Obesity is one of the main worldwide public health concerns whose clinical management demands new therapeutic approaches. Bariatric surgery is the most efficient treatment when other therapies have previously failed. Due to the role of gut microbiota in obesity development, the knowledge of the link between bariatric surgery and gut microbiota could elucidate new mechanistic approaches. This study aims to evaluate the long-term effects of bariatric surgery in the faecal metagenome and metabolome of patients with severe obesity. Faecal and blood samples were collected before and four years after the intervention from patients with severe obesity. Biochemical, metagenomic and metabolomic analyses were performed and faecal short-chain fatty acids were measured. Bariatric surgery improved the obesity-related status of patients and significantly reshaped gut microbiota composition. Moreover, this procedure was associated with a specific metabolome profile characterized by a reduction in energetic and amino acid metabolism. Acetate, butyrate and propionate showed a significant reduction with bariatric surgery. Finally, correlation analysis suggested the existence of a long-term compositional and functional gut microbiota profile associated with the intervention. In conclusion, bariatric surgery triggered long-lasting effects on gut microbiota composition and faecal metabolome that could be associated with the remission of obesity.


Subject(s)
Bariatric Surgery , Feces/chemistry , Gastrointestinal Microbiome , Metabolome , Obesity, Morbid/microbiology , Obesity, Morbid/surgery , DNA/analysis , Fatty Acids, Volatile/analysis , Feces/microbiology , Humans , Longitudinal Studies , Metagenomics , Weight Loss
7.
Nutrients ; 13(1)2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33374578

ABSTRACT

Gut microbiota (GM) is involved in the maintenance of physiological homeostasis, thus the alteration of its composition and functionality has been associated with many pathologies such as metabolic diseases, and could also be linked with the progressive degenerative process in aging. Nowadays, life expectancy is continuously rising, so the number of elder people and the consequent related pathologies demand new strategies to achieve healthy aging. Besides, actual lifestyle patterns make metabolic diseases a global epidemic with increasing trends, responsible for a large mortality and morbidity in adulthood and also compromising the health status of later stages of life. Metabolic diseases and aging share a profile of low-grade inflammation and innate immunity activation, which may have disturbances of GM composition as the leading mechanism. Thus, GM emerges as a therapeutic target with a double impact in the elderly, counteracting both aging itself and the frequent metabolic diseases in this population. This review summarizes the role and compositional changes of the GM in aging and its modulation through nutritional interventions and physical exercise as a strategy to counteract the aging process and the related metabolic diseases.


Subject(s)
Aging , Exercise Therapy , Gastrointestinal Microbiome , Metabolic Diseases/therapy , Nutrition Therapy , Aged , Aging/physiology , Exercise Therapy/methods , Gastrointestinal Microbiome/physiology , Humans , Metabolic Diseases/diet therapy , Nutrition Therapy/methods , Probiotics/therapeutic use
8.
Exp Mol Med ; 52(7): 1048-1061, 2020 07.
Article in English | MEDLINE | ID: mdl-32624568

ABSTRACT

Childhood obesity has reached epidemic levels and is a serious health concern associated with metabolic syndrome, nonalcoholic fatty liver disease, and gut microbiota alterations. Physical exercise is known to counteract obesity progression and modulate the gut microbiota composition. This study aims to determine the effect of a 12-week strength and endurance combined training program on gut microbiota and inflammation in obese pediatric patients. Thirty-nine obese children were assigned randomly to the control or training group. Anthropometric and biochemical parameters, muscular strength, and inflammatory signaling pathways in mononuclear cells were evaluated. Bacterial composition and functionality were determined by massive sequencing and metabolomic analysis. Exercise reduced plasma glucose levels and increased dynamic strength in the upper and lower extremities compared with the obese control group. Metagenomic analysis revealed a bacterial composition associated with obesity, showing changes at the phylum, class, and genus levels. Exercise counteracted this profile, significantly reducing the Proteobacteria phylum and Gammaproteobacteria class. Moreover, physical activity tended to increase some genera, such as Blautia, Dialister, and Roseburia, leading to a microbiota profile similar to that of healthy children. Metabolomic analysis revealed changes in short-chain fatty acids, branched-chain amino acids, and several sugars in response to exercise, in correlation with a specific microbiota profile. Finally, the training protocol significantly inhibited the activation of the obesity-associated NLRP3 signaling pathway. Our data suggest the existence of an obesity-related deleterious microbiota profile that is positively modified by physical activity intervention. Exercise training could be considered an efficient nonpharmacological therapy, reducing inflammatory signaling pathways induced by obesity in children via microbiota modulation.


Subject(s)
Exercise/physiology , Gastrointestinal Microbiome , Inflammation/microbiology , Pediatric Obesity/metabolism , Pediatric Obesity/microbiology , Pediatric Obesity/physiopathology , Signal Transduction , Case-Control Studies , Child , Endurance Training , Female , Humans , Male , Metabolomics , Pediatric Obesity/blood , Phylogeny , Principal Component Analysis
9.
Mol Nutr Food Res ; 63(20): e1900487, 2019 10.
Article in English | MEDLINE | ID: mdl-31322321

ABSTRACT

SCOPE: Gut microbiota contributes to non-alcoholic fatty liver disease (NAFLD) pathogenesis by multiple mechanisms not yet completely understood. Novel differential features between germ-free mice (GFm) transplanted with protective or non-protective cecal microbiota against NAFLD are investigated. METHODS AND RESULTS: Gut microbiota composition, plasma, and fecal bile acids (BAs) and liver mRNAs are quantified in GFm recipients from four donor mice differing in NAFLD severity (control diet, high-fat diet [HFD]-responder, HFD-non-responder, and quercetin-supplemented HFD). Transplanted GFm are on control or HFD for 16-weeks. Multivariate analysis shows that GFm colonized with microbiota from HFD-non-responder and quercetin supplemented-HFD donors (protected against NAFLD) clusters together, whereas GFm colonized with microbiota from control and HFD-responder mice (non-protected against NAFLD) establishes another cluster. Protected phenotype is associated with increased gut Desulfovibrio and Oscillospira, reduced gut Bacteroides and Oribacterium, lower primary and higher secondary BAs in plasma and feces, induction of hepatic BA transporters, and repression of hepatic lipogenic and BA synthesis genes. CONCLUSION: Protective gut microbiota associates with increased specific secondary BAs, which likely inhibit lipogenic pathways and enhance bile flow in the liver. This novel cross-talk between gut and liver, via plasma BAs, that promotes protection against NAFLD may have clinical and nutritional relevance.


Subject(s)
Bile Acids and Salts/blood , Gastrointestinal Microbiome , Liver/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , Animals , Diet, High-Fat , Ethanol/blood , Male , Mice , Mice, Inbred C57BL , Transcriptome
10.
Dis Model Mech ; 12(5)2019 04 30.
Article in English | MEDLINE | ID: mdl-30971408

ABSTRACT

Childhood obesity has reached epidemic levels, representing one of the most serious public health concerns associated with metabolic syndrome and non-alcoholic fatty liver disease (NAFLD). There is limited clinical experience concerning pediatric NAFLD patients, and thus the therapeutic options are scarce. The aim of this study was to evaluate the benefits of exercise on gut microbiota composition and functionality balance, and consequent effects on early obesity and NAFLD onset in an in vivo model. Juvenile (21-day-old) male Wistar rats fed a control diet or a high-fat diet (HFD) were subjected to a combined aerobic and resistance training protocol. Fecal microbiota was sequenced by an Illumina MiSeq system, and parameters related to metabolic syndrome, fecal metabolome, intestinal barrier integrity, bile acid metabolism and transport, and alteration of the gut-liver axis were measured. Exercise decreased HFD-induced body weight gain, metabolic syndrome and hepatic steatosis, as a result of its lipid metabolism modulatory capacity. Gut microbiota composition and functionality were substantially modified as a consequence of diet, age and exercise intervention. In addition, the training protocol increased Parabacteroides, Bacteroides and Flavobacterium genera, correlating with a beneficial metabolomic profile, whereas Blautia, Dysgonomonas and Porphyromonas showed an opposite pattern. Exercise effectively counteracted HFD-induced microbial imbalance, leading to intestinal barrier preservation, which, in turn, prevented deregulation of the gut-liver axis and improved bile acid homeostasis, determining the clinical outcomes of NAFLD. In conclusion, we provide scientific evidence highlighting the benefits of gut microbiota composition and functionality modulation by physical exercise protocols in the management of early obesity and NAFLD development.


Subject(s)
Gastrointestinal Microbiome , Intestines/pathology , Liver/pathology , Non-alcoholic Fatty Liver Disease/microbiology , Obesity/microbiology , Physical Conditioning, Animal , Animals , Bile Acids and Salts/metabolism , Diet, High-Fat , Disease Models, Animal , Enterohepatic Circulation , Fatty Acids/biosynthesis , Feces , Inflammation/pathology , Lipid Metabolism , Liver/injuries , Male , Metabolic Syndrome/complications , Metabolome , Oxidative Stress , Principal Component Analysis , Rats, Wistar
11.
Rev. esp. enferm. dig ; 111(4): 275-282, abr. 2019. tab, graf
Article in English | IBECS | ID: ibc-189924

ABSTRACT

Introduction: increasing evidence suggests a role of intestinal dysbiosis in obesity and non-alcoholic fatty liver disease (NAFLD). The advances in recent years with regard to the role of the gut microbiota raise the potential utility of new therapeutic approaches based on the modification of the microbiome. Objective: the aim of this study was to compare the bacterial communities in obese patients with or without NAFLD to those of healthy controls. Patients and methods: the fecal microbiota composition of 20 healthy adults, 36 obese patients with NAFLD and 17 obese patients without NAFLD was determined by 16S ribosomal RNA sequencing using the Illumina MiSeq system. Results: the results highlighted significant differences in the phylum Firmicutes between patients with and without NAFLD, which was a determining factor of the disease and supported its possible role as a marker of NAFLD. At the genus level, the relative abundance of Blautia, Alkaliphilus, Flavobacterium and Akkermansia was reduced in obese patients, both with or without NAFLD, compared to healthy controls. Furthermore, the number of sequences from the genus Streptococcus was significantly higher in patients with NAFLD in comparison with individuals without the disease, constituting another possible marker. Comparison of bacterial communities at the genus level by a principal coordinate analysis indicated that the bacterial communities of patients with NAFLD were dispersed and did not form a group. Conclusion: in conclusion, these results indicate the role of intestinal dysbiosis in the development of NAFLD associated with obesity. There was a differential microbiota profile between obese patients, with and without NAFLD. Thus, supporting gut microbiota modulation as a therapeutic alternative for the prevention and treatment of NAFLD


No disponible


Subject(s)
Humans , Male , Female , Adult , Middle Aged , Gastrointestinal Microbiome/immunology , Non-alcoholic Fatty Liver Disease/physiopathology , Fatty Liver/physiopathology , Dysbiosis/microbiology , Obesity/physiopathology , Host Microbial Interactions/immunology , Fecal Microbiota Transplantation , Bacteroidetes/isolation & purification , Case-Control Studies , RNA, Ribosomal, 16S/immunology , High-Throughput Nucleotide Sequencing/methods , Proteobacteria/isolation & purification , Metabolic Syndrome/physiopathology
12.
Plant Dis ; 103(6): 1181-1188, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30908127

ABSTRACT

Epidemics of tomato yellow leaf curl disease (TYLCD) caused by tomato yellow leaf curl-like begomoviruses (genus Begomovirus, family Geminiviridae) severely damage open field and protected tomato crops worldwide. Intensive application of insecticides against the whitefly vector Bemisia tabaci is generally used as control strategy to reduce TYLCD impact. This practice, however, is frequently ineffective and has a negative impact on the environment and human health. TYLCD-resistant varieties are commercially available, but cultivation of susceptible traditional tasting ones is also requested if possible. For susceptible tomatoes, here we show that using whitefly optical barriers by means of UV-blocking plastics in protected crops can contribute to reducing TYLCD damage and increasing commercial fruit yield. Moreover, induction of systemic acquired resistance by application of the elicitor of plant defense acibenzolar-S-methyl was effective to reduce yield losses when viral pressure was moderate. Interestingly, combining both practices in protected tomato crops can result in a significant TYLCD control. Therefore, these control practices are proposed to be used commercially as management alternatives to include in integrated management of TYLCD.


Subject(s)
Begomovirus , Disease Resistance , Hemiptera , Solanum lycopersicum , Animals , Begomovirus/physiology , Crops, Agricultural/virology , Hemiptera/virology , Solanum lycopersicum/virology
13.
Rev Esp Enferm Dig ; 111(4): 275-282, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30810328

ABSTRACT

INTRODUCTION: increasing evidence suggests a role of intestinal dysbiosis in obesity and non-alcoholic fatty liver disease (NAFLD). The advances in recent years with regard to the role of the gut microbiota raise the potential utility of new therapeutic approaches based on the modification of the microbiome. OBJECTIVE: the aim of this study was to compare the bacterial communities in obese patients with or without NAFLD to those of healthy controls. PATIENTS AND METHODS: the fecal microbiota composition of 20 healthy adults, 36 obese patients with NAFLD and 17 obese patients without NAFLD was determined by 16S ribosomal RNA sequencing using the Illumina MiSeq system. RESULTS: the results highlighted significant differences in the phylum Firmicutes between patients with and without NAFLD, which was a determining factor of the disease and supported its possible role as a marker of NAFLD. At the genus level, the relative abundance of Blautia, Alkaliphilus, Flavobacterium and Akkermansia was reduced in obese patients, both with or without NAFLD, compared to healthy controls. Furthermore, the number of sequences from the genus Streptococcus was significantly higher in patients with NAFLD in comparison with individuals without the disease, constituting another possible marker. Comparison of bacterial communities at the genus level by a principal coordinate analysis indicated that the bacterial communities of patients with NAFLD were dispersed and did not form a group. CONCLUSION: in conclusion, these results indicate the role of intestinal dysbiosis in the development of NAFLD associated with obesity. There was a differential microbiota profile between obese patients, with and without NAFLD. Thus, supporting gut microbiota modulation as a therapeutic alternative for the prevention and treatment of NAFLD.


Subject(s)
Dysbiosis/microbiology , Feces/microbiology , Gastrointestinal Microbiome , Non-alcoholic Fatty Liver Disease/microbiology , Obesity/microbiology , Adult , Bacterial Load , Bacteroidetes/isolation & purification , Case-Control Studies , Female , Firmicutes/isolation & purification , Humans , Male , Metabolic Syndrome/diagnosis , Middle Aged , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/pathology , Obesity/complications , Polymerase Chain Reaction , Proteobacteria/isolation & purification
14.
Mol Nutr Food Res ; 63(8): e1800930, 2019 04.
Article in English | MEDLINE | ID: mdl-30680920

ABSTRACT

SCOPE: Modulation of intestinal microbiota has emerged as a new therapeutic approach for non-alcoholic fatty liver disease (NAFLD). Herein, it is addressed whether gut microbiota modulation by quercetin and intestinal microbiota transplantation can influence NAFLD development. METHODS AND RESULTS: Gut microbiota donor mice are selected according to their response to high-fat diet (HFD) and quercetin in terms of obesity and NAFLD-related biomarkers. Germ-free recipients displayed metabolic phenotypic differences derived from interactions between microbiota transplanted, diets, and quercetin. Based on the evaluation of hallmark characteristics of NAFLD, it is found that gut microbiota transplantation from the HFD-non-responder donor and the HFD-fed donor with the highest response to quercetin results in a protective phenotype against HFD-induced NAFLD, in a mechanism that involves gut-liver axis alteration blockage in these receivers. Gut microbiota from the HFD-responder donor predisposed transplanted germ-free mice to NAFLD. Divergent protective and deleterious metabolic phenotypes exhibited are related to definite microbial profiles in recipients, highlighting the predominant role of Akkermansia genus in the protection from obesity-associated NAFLD development. CONCLUSIONS: The results provide scientific support for the prebiotic capacity of quercetin and the transfer of established metabolic profiles through gut microbiota transplantation as a protective strategy against the development of obesity-related NAFLD.


Subject(s)
Diet, High-Fat/adverse effects , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Non-alcoholic Fatty Liver Disease/prevention & control , Quercetin/pharmacology , Animals , Endotoxemia/etiology , Endotoxemia/therapy , Fatty Acids, Volatile/metabolism , Inflammasomes , Insulin Resistance , Liver/drug effects , Liver/pathology , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/microbiology , Obesity/complications , Obesity/drug therapy , Obesity/microbiology , Verrucomicrobia/physiology
15.
Viruses ; 11(1)2019 01 09.
Article in English | MEDLINE | ID: mdl-30634476

ABSTRACT

The tomato leaf curl disease (TYLCD) is associated with infections of several species of begomoviruses (genus Begomovirus, family Geminiviridae) and causes severe damage to tomatoes throughout tropical and sub-tropical regions of the world. Among others, the Tomato yellow leaf curl Sardinia virus (TYLCSV) species causes damage in the Mediterranean Basin since early outbreaks occurred. Nevertheless, scarce information is available about the diversity of TYLCSV. Here, we study this aspect based on the sequence information accessible in databases. Isolates of two taxonomically differentiated TYLCSV strains can be found in natural epidemics. Their evolution is mostly associated with mutation combined with selection and random genetic drift and also with inter-species recombination which is frequent in begomoviruses. Moreover, a novel putative inter-strain recombinant is reported. Although no significantly new biological behaviour was observed for this latter recombinant, its occurrence supports that as shown for other related begomoviruses, recombination continues to play a central role in the evolution of TYLCD-associated viruses and the dynamism of their populations. The confrontation of resistant tomatoes with isolates of different TYLCD-associated viruses including the novel recombinant demonstrates the existence of a variable virus x plant genotype interaction. This has already been observed for other TYLCD-associated viruses and is a challenge for the control of their impact on tomato production.


Subject(s)
Begomovirus/genetics , Evolution, Molecular , Genetic Variation , Mutation , Recombination, Genetic , Genotype , Solanum lycopersicum/virology , Phylogeny , Plant Diseases/virology
16.
Front Plant Sci ; 9: 932, 2018.
Article in English | MEDLINE | ID: mdl-30013589

ABSTRACT

Geminiviruses (family Geminiviridae) possess single-stranded circular DNA genomes that are replicated by cellular polymerases in plant host cell nuclei. In their hosts, geminivirus populations behave as ensembles of mutant and recombinant genomes, known as viral quasispecies. This favors the emergence of new geminiviruses with altered host range, facilitating new or more severe diseases or overcoming resistance traits. In warm and temperate areas several whitefly-transmitted geminiviruses of the genus Begomovirus cause the tomato yellow leaf curl disease (TYLCD) with significant economic consequences. TYLCD is frequently controlled in commercial tomatoes by using the dominant Ty-1 resistance gene. Over a 45 day period we have studied the diversification of three begomoviruses causing TYLCD: tomato yellow leaf curl virus (TYLCV), tomato yellow leaf curl Sardinia virus (TYLCSV) and tomato yellow leaf curl Malaga virus (TYLCMaV, a natural recombinant between TYLCV and TYLCSV). Viral quasispecies resulting from inoculation of geminivirus infectious clones were examined in plants of susceptible tomato (ty-1/ty-1), heterozygous resistant tomato (Ty-1/ty-1), common bean, and the wild reservoir Solanum nigrum. Differences in virus fitness across hosts were observed while viral consensus sequences remained invariant. However, the complexity and heterogeneity of the quasispecies were high, especially in common bean and the wild host. Interestingly, the presence or absence of the Ty-1 allele in tomato did not lead to differences in begomovirus mutant spectra. However, the fitness decrease of TYLCSV and TYLCV in tomato at 45 dpi might be related to an increase in CP (Coat protein) mutation frequency. In Solanum nigrum the recombinant TYLCMaV, which showed lower fitness than TYLCSV, at 45 dpi actively explored Rep (Replication associated protein) ORF but not the overlapping C4. Our results underline the importance of begomovirus mutant spectra during infections. This is especially relevant in the wild reservoir of the viruses, which has the potential to maintain highly diverse mutant spectra without modifying their consensus sequences.

17.
Curr Med Chem ; 25(7): 814-838, 2018.
Article in English | MEDLINE | ID: mdl-28925866

ABSTRACT

BACKGROUND: Autophagy is a cellular pathway with the ability to maintain cell homeostasis through the elimination of damaged or useless cellular components, and its deregulation may initiate or aggravate different human diseases. Flavonoids, a group of plant metabolites, are able to modulate different molecular and cellular processes including autophagy. OBJECTIVE: To review the effects of flavonoids on autophagy pathway in both invasive and noninvasive human diseases, focusing on the global outcomes in their progression. Moreover, the efficacy of the combination of flavonoids with drugs or other natural nontoxic compounds was also reviewed. METHODS: A literature search was performed to identify and analyze peer-reviewed publications containing in vitro and in vivo studies focused on autophagy deregulation in different proliferative and non-proliferative pathologies and the potential protective effects of flavonoids. RESULTS: Analyzed publications indicated that imbalance between cell death and survival induced by changes in autophagy play an important role in the pathophysiology of a number of human diseases. The use of different flavonoids as autophagy modulators, alone or in combination with other molecules, might be a worthy strategy in the treatment of cancer, neurodegenerative disorders, cardiovascular diseases, hepatic diseases, leishmaniasis, influenza, gastric ulcers produced by Helicobacter pylori infection, diabetes, asthma, age-related macular degeneration or osteoporosis. CONCLUSION: Flavonoids could potentially constitute important adjuvant agents of conventional therapies in the treatment of autophagy deregulation-related diseases. Moreover, combined therapy may help to diminish the doses of those conventional treatments, leading to reduced drug-derivative side effects and to improved patients' survival.


Subject(s)
Autophagy/drug effects , Flavonoids/pharmacology , Neoplasms/drug therapy , Asthma/drug therapy , Asthma/pathology , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/pathology , Cell Proliferation/drug effects , Diabetes Mellitus/drug therapy , Diabetes Mellitus/pathology , Flavonoids/chemistry , Humans , Influenza, Human/drug therapy , Influenza, Human/pathology , Leishmaniasis/drug therapy , Leishmaniasis/pathology , Liver Diseases/drug therapy , Liver Diseases/pathology , Neoplasms/pathology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/pathology , Osteoporosis/drug therapy , Osteoporosis/pathology , Stomach Ulcer/drug therapy , Stomach Ulcer/pathology
18.
Front Physiol ; 9: 1813, 2018.
Article in English | MEDLINE | ID: mdl-30618824

ABSTRACT

Obesity and associated comorbidities, including non-alcoholic fatty liver disease (NAFLD), are a major concern to public well-being worldwide due to their high prevalence among the population, and its tendency on the rise point to as important threats in the future. Therapeutic approaches for obesity-associated disorders have been circumscribed to lifestyle modifications and pharmacological therapies have demonstrated limited efficacy. Over the last few years, different studies have shown a significant role of intestinal microbiota (IM) on obesity establishment and NAFLD development. Therefore, modulation of IM emerges as a promising therapeutic strategy for obesity-associated diseases. Administration of prebiotic and probiotic compounds, fecal microbiota transplantation (FMT) and exercise protocols have shown a modulatory action over the IM. In this review we provide an overview of current approaches targeting IM which have shown their capacity to counteract NAFLD and metabolic syndrome features in human patients and animal models.

19.
Free Radic Biol Med ; 102: 188-202, 2017 01.
Article in English | MEDLINE | ID: mdl-27890642

ABSTRACT

Gut microbiota is involved in obesity, metabolic syndrome and the progression of nonalcoholic fatty liver disease (NAFLD). It has been recently suggested that the flavonoid quercetin may have the ability to modulate the intestinal microbiota composition, suggesting a prebiotic capacity which highlights a great therapeutic potential in NAFLD. The present study aims to investigate benefits of experimental treatment with quercetin on gut microbial balance and related gut-liver axis activation in a nutritional animal model of NAFLD associated to obesity. C57BL/6J mice were challenged with high fat diet (HFD) supplemented or not with quercetin for 16 weeks. HFD induced obesity, metabolic syndrome and the development of hepatic steatosis as main hepatic histological finding. Increased accumulation of intrahepatic lipids was associated with altered gene expression related to lipid metabolism, as a result of deregulation of their major modulators. Quercetin supplementation decreased insulin resistance and NAFLD activity score, by reducing the intrahepatic lipid accumulation through its ability to modulate lipid metabolism gene expression, cytochrome P450 2E1 (CYP2E1)-dependent lipoperoxidation and related lipotoxicity. Microbiota composition was determined via 16S ribosomal RNA Illumina next-generation sequencing. Metagenomic studies revealed HFD-dependent differences at phylum, class and genus levels leading to dysbiosis, characterized by an increase in Firmicutes/Bacteroidetes ratio and in Gram-negative bacteria, and a dramatically increased detection of Helicobacter genus. Dysbiosis was accompanied by endotoxemia, intestinal barrier dysfunction and gut-liver axis alteration and subsequent inflammatory gene overexpression. Dysbiosis-mediated toll-like receptor 4 (TLR-4)-NF-κB signaling pathway activation was associated with inflammasome initiation response and reticulum stress pathway induction. Quercetin reverted gut microbiota imbalance and related endotoxemia-mediated TLR-4 pathway induction, with subsequent inhibition of inflammasome response and reticulum stress pathway activation, leading to the blockage of lipid metabolism gene expression deregulation. Our results support the suitability of quercetin as a therapeutic approach for obesity-associated NAFLD via its anti-inflammatory, antioxidant and prebiotic integrative response.


Subject(s)
Metabolic Syndrome/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Obesity/drug therapy , Quercetin/administration & dosage , Toll-Like Receptor 4/metabolism , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Humans , Insulin Resistance/genetics , Intestines/microbiology , Lipid Metabolism/genetics , Liver/metabolism , Liver/pathology , Metabolic Syndrome/metabolism , Metabolic Syndrome/microbiology , Metabolic Syndrome/pathology , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/microbiology , Non-alcoholic Fatty Liver Disease/pathology , Obesity/metabolism , Obesity/microbiology , Obesity/pathology , Signal Transduction/drug effects , Toll-Like Receptor 4/genetics
20.
Viruses ; 8(11)2016 11 10.
Article in English | MEDLINE | ID: mdl-27834936

ABSTRACT

Tomato leaf curl New Delhi virus (ToLCNDV) is a whitefly-transmitted bipartite begomovirus (genus Begomovirus, family Geminiviridae) that causes damage to multiple cultivated plant species mainly belonging to the Solanaceae and Cucurbitaceae families. ToLCNDV was limited to Asian countries until 2012, when it was first reported in Spain, causing severe epidemics in cucurbit crops. Here, we show that a genetically-uniform ToLCNDV population is present in Spain, compatible with a recent introduction. Analyses of ToLCNDV isolates reported from other parts of the world indicated that this virus has a highly heterogeneous population genetically with no evident geographical, plant host or year-based phylogenetic groups observed. Isolates emerging in Spain belong to a strain that seems to have evolved by recombination. Isolates of this strain seem adapted to infecting cucurbits, but poorly infect tomatoes.


Subject(s)
Begomovirus/classification , Begomovirus/isolation & purification , Phylogeny , Plant Diseases/virology , Begomovirus/genetics , Cucurbitaceae/virology , Evolution, Molecular , Recombination, Genetic , Spain
SELECTION OF CITATIONS
SEARCH DETAIL
...