Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Sep Sci ; 45(18): 3431-3442, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35855656

ABSTRACT

Naoling Pian is a prescription composed of 15 herbs, which is mainly used for the treatment of insomnia in clinical practice. However, the chemical constituents in Naoling Pian are numerous and unclear, which hinders the interpretation of its bioactive constituents and the subsequent research on the material basis for pharmacodynamics. The purpose of this study is to develop a rapid method for identifying the chemical constituents of Naoling Pian using high-throughput ultra-performance liquid chromatography quadrupole time of flight coupled with mass spectrometry combined with a software platform for data processing. The whole composition of Naoling Pian was characterized in positive and negative ion modes. In this experiment, an overall total of 201 constituents were identified by using reference standards, online and self-built databases matching, fragmentation rules analysis of mass spectrometry peaks with a software platform. Meanwhile, Naoling Pian was analyzed for the first time using liquid chromatography-mass spectrometry method, the constituents could be identified in a quick and accurate manner, and the results could provide a scientific basis for the follow-up research on the pharmacodynamic material basis and quality control of Naoling Pian.


Subject(s)
Drugs, Chinese Herbal , Tandem Mass Spectrometry , Chromatography, High Pressure Liquid/methods , Chromatography, Liquid , Drugs, Chinese Herbal/analysis , Reference Standards , Tandem Mass Spectrometry/methods
2.
Sci Rep ; 11(1): 3534, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33574421

ABSTRACT

Human milk is considered the optimal nutrition for infants and found to contain significant numbers of viable bacteria. The aim of the study was to assess the effects of a specific synbiotic combination at doses closer to the bacterial cells present in human milk, on intestinal bifidobacteria proportions (relative abundance), reduction of potential pathogens and gut physiological conditions. A clinical study was conducted in 290 healthy infants aged from 6 to 19 weeks. Infants received either a control infant formula or one of the two investigational infant formulas (control formula with 0.8 g/100 ml scGOS/lcFOS and Bifidobacterium breve M-16V at either 1 × 104 cfu/ml or 1 × 106 cfu/ml). Exclusively breastfed infants were included as a reference. Analyses were performed on intention-to-treat groups and all-subjects-treated groups. After 6 weeks of intervention, the synbiotics at two different doses significantly increased the bifidobacteria proportions in healthy infants. The synbiotic supplementation also decreased the prevalence (infants with detectable levels) and the abundance of C. difficile. Closer to the levels in the breastfed reference group, fecal pH was significantly lower while L-lactate concentrations and acetate proportions were significantly higher in the synbiotic groups. All formulas were well tolerated and all groups showed a comparable safety profile based on the number and severity of adverse events and growth. In healthy infants, supplementation of infant-type bifidobacterial strain B. breve M-16V, at a dose close to bacterial numbers found in human milk, with scGOS/lcFOS (9:1) created a gut environment closer to the breastfed reference group. This specific synbiotic mixture may also support gut microbiota resilience during early life.Clinical Trial Registration This clinical study named Color Synbiotics Study, was registered in ClinicalTrials.gov on 18 March 2013. Registration number is NCT01813175. https://clinicaltrials.gov/ct2/show/NCT01813175 .


Subject(s)
Bacterial Infections/prevention & control , Bifidobacterium/isolation & purification , Clostridioides difficile/isolation & purification , Milk, Human/microbiology , Synbiotics/administration & dosage , Bacterial Infections/microbiology , Bifidobacterium/metabolism , Bifidobacterium breve/isolation & purification , Bifidobacterium breve/metabolism , Breast Feeding , Clostridioides difficile/pathogenicity , Double-Blind Method , Feces/microbiology , Female , Gastrointestinal Microbiome/drug effects , Humans , Infant , Infant Formula/microbiology , Infant, Newborn , Male
3.
Curr Biol ; 30(19): 3761-3774.e6, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32822606

ABSTRACT

Birth by Caesarean (C)-section impacts early gut microbiota colonization and is associated with an increased risk of developing immune and metabolic disorders. Moreover, alterations of the microbiome have been shown to affect neurodevelopmental trajectories. However, the long-term effects of C-section on neurobehavioral processes remain unknown. Here, we demonstrated that birth by C-section results in marked but transient changes in microbiome composition in the mouse, in particular, the abundance of Bifidobacterium spp. was depleted in early life. Mice born by C-section had enduring social, cognitive, and anxiety deficits in early life and adulthood. Interestingly, we found that these specific behavioral alterations induced by the mode of birth were also partially corrected by co-housing with vaginally born mice. Finally, we showed that supplementation from birth with a Bifidobacterium breve strain, or with a dietary prebiotic mixture that stimulates the growth of bifidobacteria, reverses selective behavioral alterations in C-section mice. Taken together, our data link the gut microbiota to behavioral alterations in C-section-born mice and suggest the possibility of developing adjunctive microbiota-targeted therapies that may help to avert long-term negative consequences on behavior associated with C-section birth mode.


Subject(s)
Cesarean Section/adverse effects , Gastrointestinal Microbiome/physiology , Nervous System Diseases/microbiology , Animals , Bifidobacterium/growth & development , Bifidobacterium/metabolism , Cesarean Section/psychology , Disease Models, Animal , Feces/microbiology , Female , Mice , Pregnancy
4.
Neurosci Biobehav Rev ; 95: 191-201, 2018 12.
Article in English | MEDLINE | ID: mdl-30195933

ABSTRACT

In the first 2-3 years of life, the gut microbiota of infants quickly becomes diverse and rich. Disruptions in the evolving gut microbiota during this critical developmental period can impact brain development. Communication between the microbiota, gut and brain is driven by hormonal and neural regulation, as well as immune and metabolic pathways, however, our understanding of how the parallel developments that may underlie this communication are limited. In this paper, we review the known associations between the gut microbiota and brain development and brain function in early life, speculate on the potential mechanisms involved in this complex relationship and describe how nutritional intervention can further modulate the microbiota and, ultimately, brain development and function.


Subject(s)
Brain/growth & development , Gastrointestinal Microbiome , Animals , Brain/microbiology , Brain/physiology , Humans
5.
Nutr Rev ; 75(4): 225-240, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28379454

ABSTRACT

Microbial colonization of the gastrointestinal tract is an essential process that modulates host physiology and immunity. Recently, researchers have begun to understand how and when these microorganisms colonize the gut and the early-life factors that impact their natural ecological establishment. The vertical transmission of maternal microbes to the offspring is a critical factor for host immune and metabolic development. Increasing evidence also points to a role in the wiring of the gut-brain axis. This process may be altered by various factors such as mode of delivery, gestational age at birth, the use of antibiotics in early life, infant feeding, and hygiene practices. In fact, these early exposures that impact the intestinal microbiota have been associated with the development of diseases such as obesity, type 1 diabetes, asthma, allergies, and even neurodevelopmental disorders. The present review summarizes the impact of cesarean birth on the gut microbiome and the health status of the developing infant and discusses possible preventative and restorative strategies to compensate for early-life microbial perturbations.


Subject(s)
Cesarean Section , Gastrointestinal Microbiome , Brain/growth & development , Female , Gastrointestinal Tract/microbiology , Humans , Pregnancy
6.
PLoS One ; 9(5): e97523, 2014.
Article in English | MEDLINE | ID: mdl-24830946

ABSTRACT

Colonizing commensal bacteria after birth are required for the proper development of the gastrointestinal tract. It is believed that bacterial colonization pattern in neonatal gut affects gut barrier function and immune system maturation. Studies on the development of faecal microbiota in infants showed that the neonatal gut was first colonized with enterococci followed by other microbiota such as Bifidobacterium. Other studies showed that babies who developed allergy were less often colonized with Enterococcus during the first month of life as compared to healthy infants. Many studies have been conducted to elucidate how bifidobacteria or lactobacilli, some of which are considered probiotic, regulate infant gut immunity. However, fewer studies have been focused on enterococi. In our study, we demonstrate that E. faecalis, isolated from healthy newborns, suppress inflammatory responses activated in vivo and in vitro. We found E. faecalis attenuates proinflammatory cytokine secretions, especially IL-8, through JNK and p38 signaling pathways. This finding shed light on how the first colonizer, E.faecalis, regulates inflammatory responses in the host.


Subject(s)
Enterococcus faecalis/metabolism , Inflammation/metabolism , Inflammation/microbiology , MAP Kinase Signaling System , Animals , Caco-2 Cells , Culture Media, Conditioned/chemistry , Enterococcus faecalis/genetics , HCT116 Cells , HT29 Cells , Humans , Indonesia , Infant , Infant, Newborn , Interleukin-1beta/metabolism , Interleukin-8/metabolism , Intestinal Mucosa/metabolism , Intestines/microbiology , Lactobacillus/genetics , Lactobacillus/metabolism , Male , Mice , Mice, Inbred C57BL , Phylogeny , Polymerase Chain Reaction , RNA, Ribosomal, 16S/genetics , Salmonella typhimurium/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
BMC Med Genomics ; 6: 41, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-24119614

ABSTRACT

BACKGROUND: The majority of gastric cancer cases are believed to be caused by chronic infection with the bacterium Helicobacter pylori, and atrophic corpus gastritis is a predisposing condition to gastric cancer development. We aimed to increase understanding of the molecular details of atrophy by performing a global transcriptome analysis of stomach tissue. METHODS: Biopsies from patients with different stages of H. pylori infection were taken from both the antrum and corpus mucosa and analyzed on microarrays. The stages included patients without current H. pylori infection, H. pylori-infected without corpus atrophy and patients with current or past H. pylori-infection with corpus-predominant atrophic gastritis. RESULTS: Using clustering and integrated analysis, we found firm evidence for antralization of the corpus mucosa of atrophy patients. This antralization harbored gain of gastrin expression, as well as loss of expression of corpus-related genes, such as genes associated with acid production, energy metabolism and blood clotting. The analyses provided detailed molecular evidence for simultaneous intestinal metaplasia (IM) and spasmolytic polypeptide expressing metaplasia (SPEM) in atrophic corpus tissue. Finally, acidic mammalian chitinase, a chitin-degrading enzyme produced by chief cells, was shown to be strongly down-regulated in corpus atrophy. CONCLUSIONS: Transcriptome analysis revealed several gene groups which are related to development of corpus atrophy, some of which were increased also in H. pylori-infected non-atrophic patients. Furthermore, loss of acidic chitinase expression is a promising marker for corpus atrophy.


Subject(s)
Chitinases/genetics , Gastric Mucosa/microbiology , Gastritis, Atrophic/enzymology , Gastritis, Atrophic/genetics , Helicobacter pylori/physiology , Transcriptome , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , Blood Vessels/physiopathology , Chitinases/deficiency , Energy Metabolism/genetics , Female , Gastric Mucosa/blood supply , Gastric Mucosa/metabolism , Gastritis, Atrophic/metabolism , Gastritis, Atrophic/physiopathology , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Transcription, Genetic
8.
Proc Natl Acad Sci U S A ; 108(7): 3047-52, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21282636

ABSTRACT

Microbial colonization of mammals is an evolution-driven process that modulate host physiology, many of which are associated with immunity and nutrient intake. Here, we report that colonization by gut microbiota impacts mammalian brain development and subsequent adult behavior. Using measures of motor activity and anxiety-like behavior, we demonstrate that germ free (GF) mice display increased motor activity and reduced anxiety, compared with specific pathogen free (SPF) mice with a normal gut microbiota. This behavioral phenotype is associated with altered expression of genes known to be involved in second messenger pathways and synaptic long-term potentiation in brain regions implicated in motor control and anxiety-like behavior. GF mice exposed to gut microbiota early in life display similar characteristics as SPF mice, including reduced expression of PSD-95 and synaptophysin in the striatum. Hence, our results suggest that the microbial colonization process initiates signaling mechanisms that affect neuronal circuits involved in motor control and anxiety behavior.


Subject(s)
Brain/metabolism , Exploratory Behavior/physiology , Gastrointestinal Tract/microbiology , Maze Learning/physiology , Motor Activity/physiology , Analysis of Variance , Animals , Chromatography, High Pressure Liquid , Disks Large Homolog 4 Protein , Electrophoresis, Polyacrylamide Gel , Germ-Free Life , Guanylate Kinases , In Situ Hybridization , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/metabolism , Mice , Microarray Analysis , Polymerase Chain Reaction , Sequence Analysis, RNA , Specific Pathogen-Free Organisms , Synaptophysin/metabolism
9.
Proc Natl Acad Sci U S A ; 105(6): 1943-8, 2008 Feb 12.
Article in English | MEDLINE | ID: mdl-18234854

ABSTRACT

The postembryonic development of the gastrointestinal tract is subject to regulation by the colonizing microbiota. This maturation process requires the commensal bacteria to cross-talk with host cells by way of recognizing receptors and inducing signaling pathways to activate transcription factors such as the nuclear receptors. Here, we show that in colonic cell lines and in primary colonic cells, Enterococcus faecalis isolated from newborn babies possess the ability to regulate peroxisome proliferator-activated receptor-gamma1 (PPARgamma1) activity through phosphorylation. This results in elevated DNA binding and transcriptional activation of downstream target genes, including IL-10, a cytokine known to modulate innate immune function. Furthermore, phosphorylation appears tightly regulated as phospho-PPARgamma1 becomes an immediate substrate for degradation possibly to curtail any extended transactivation. The involvement of PPARgamma1 in a myriad of physiological processes further confirms that microflora-driven regulation might be important for a number of homeostatic strategies in the gut.


Subject(s)
Colon/metabolism , Enterococcus faecalis/physiology , Interleukin-10/metabolism , PPAR gamma/metabolism , Colon/cytology , Colon/microbiology , DNA/metabolism , Gene Expression , HT29 Cells , Humans , Infant, Newborn , Interleukin-10/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Ligands , Membrane Proteins/genetics , Membrane Proteins/metabolism , Perilipin-2 , Phosphorylation , Protein Binding
10.
World J Gastroenterol ; 14(7): 1067-76, 2008 Feb 21.
Article in English | MEDLINE | ID: mdl-18286689

ABSTRACT

AIM: To investigate the ability of Lactic acid bacteria (LAB) to modulate inflammatory reaction in human intestinal cell lines (Caco-2, HT-29 and HCT116). Different strains of LAB isolated from new born infants and fermented milk, together with the strains obtained from culture collections were tested. METHODS: LABs were treated with human intestinal cell lines. ELISA was used to detect IL-8 and TGF-beta protein secretion. Cytokines and Toll like receptors (TLRs) gene expression were assessed using RT-PCR. Conditional medium, sonicated bacteria and UV killed bacteria were used to find the effecter molecules on the bacteria. Carbohydrate oxidation and protein digestion were applied to figure out the molecules' residues. Adhesion assays were further carried out. RESULTS: It was found that Enterococcus faecalis is the main immune modulator among the LABs by downregulation of IL-8 secretion and upregulation of TGF-beta. Strikingly, the effect was only observed in four strains of E. faecalis out of the 27 isolated and tested. This implies strain dependent immunomodulation in the host. In addition, E. faecalis may regulate inflammatory responses through TLR3, TLR4, TLR9 and TRAF6. Carbohydrates on the bacterial cell surface are involved in both its adhesion to intestinal cells and regulation of inflammatory responses in the host. CONCLUSION: These data provide a case for the modulation of intestinal mucosal immunity in which specific strains of E. faecalis have uniquely evolved to maintain colonic homeostasis and regulate inflammatory responses.


Subject(s)
Enterococcus faecalis/immunology , Inflammation/prevention & control , Intestines/immunology , Intestines/microbiology , Bacterial Adhesion , Base Sequence , Caco-2 Cells , Cell Line , Cytokines/genetics , DNA Primers/genetics , Down-Regulation , Enterococcus faecalis/isolation & purification , Humans , Immunity, Mucosal , Infant, Newborn , Inflammation/genetics , Inflammation/immunology , Inflammation/microbiology , Interleukin-8/biosynthesis , Toll-Like Receptors/genetics , Transforming Growth Factor beta/biosynthesis
11.
Zhongguo Zhong Yao Za Zhi ; 30(6): 422-6, 2005 Mar.
Article in Chinese | MEDLINE | ID: mdl-15810444

ABSTRACT

OBJECTIVE: To characterize the different varieties of Pseudostellaria heterophylla during cultivation. METHOD: Using systematic selection in the main productive areas, the techniques of random design, all varieties were observed for 3 years. RESULT: The biological and 425 productive characteristics of P. heterophylla var. macrophylla, P. heterophylla var. Foliolum, and P. heterophylla var. anvense were significantly different (P < 0.01). There were also differences in ecological adaptability, plant characteristics, pollen granule, chromosomes, and isoenzyme of the three cultivars. CONCLUSION: The strain types of P. heterophylla was denominated for the first time. The characteristics and productivity index system of P. heterophylla varieties were determined.


Subject(s)
Caryophyllaceae/anatomy & histology , Chromosomes, Plant , Lipase/analysis , Plants, Medicinal/anatomy & histology , Caryophyllaceae/enzymology , Caryophyllaceae/genetics , Catechol Oxidase/analysis , Ecosystem , Flowers/anatomy & histology , Peroxidase/analysis , Plant Leaves/anatomy & histology , Plant Roots/anatomy & histology , Plants, Medicinal/enzymology , Plants, Medicinal/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...