scispace - formally typeset
Search or ask a question
Author

Yan-Xia Zhou

Other affiliations: Shandong University
Bio: Yan-Xia Zhou is an academic researcher from Lawrence Berkeley National Laboratory. The author has contributed to research in topics: Strain (injury) & Medicine. The author has an hindex of 1, co-authored 3 publications receiving 32 citations. Previous affiliations of Yan-Xia Zhou include Shandong University.

Papers
More filters
Journal ArticleDOI
TL;DR: New evidence is provided for a link between Lactobacillus and memory and possible new avenues for treating memory impairment disorders using specific gut microbial inoculants and/or metabolites are opened.
Abstract: Recent evidence has linked the gut microbiome to host behavior via the gut–brain axis [1–3]; however, the underlying mechanisms remain unexplored. Here, we determined the links between host genetics, the gut microbiome and memory using the genetically defined Collaborative Cross (CC) mouse cohort, complemented with microbiome and metabolomic analyses in conventional and germ-free (GF) mice. A genome-wide association analysis (GWAS) identified 715 of 76,080 single-nucleotide polymorphisms (SNPs) that were significantly associated with short-term memory using the passive avoidance model. The identified SNPs were enriched in genes known to be involved in learning and memory functions. By 16S rRNA gene sequencing of the gut microbial community in the same CC cohort, we identified specific microorganisms that were significantly correlated with longer latencies in our retention test, including a positive correlation with Lactobacillus. Inoculation of GF mice with individual species of Lactobacillus (L. reuteri F275, L. plantarum BDGP2 or L. brevis BDGP6) resulted in significantly improved memory compared to uninoculated or E. coli DH10B inoculated controls. Untargeted metabolomics analysis revealed significantly higher levels of several metabolites, including lactate, in the stools of Lactobacillus-colonized mice, when compared to GF control mice. Moreover, we demonstrate that dietary lactate treatment alone boosted memory in conventional mice. Mechanistically, we show that both inoculation with Lactobacillus or lactate treatment significantly increased the levels of the neurotransmitter, gamma-aminobutyric acid (GABA), in the hippocampus of the mice. Together, this study provides new evidence for a link between Lactobacillus and memory and our results open possible new avenues for treating memory impairment disorders using specific gut microbial inoculants and/or metabolites.

56 citations

Journal ArticleDOI
TL;DR: Alginate as a preferred polymer and recent advances in alginate-based polymers for probiotics delivery systems are presented and future directions, specifically on the use of nanomaterials as well as prebiotics, are delineated to further facilitate subsequent researchers in selecting more favorable materials and technology for probiotic delivery.
Abstract: Probiotics exhibit many health benefits and a great potential for broad applications in pharmaceutical fields, such as prevention and treatment of gastrointestinal tract diseases (irritable bowel syndrome), prevention and therapy of allergies, certain anticancer effects, and immunomodulation. However, their applications are limited by the low viability and metabolic activity of the probiotics during processing, storage, and delivery in the digestive tract. To overcome the mentioned limitations, probiotic delivery systems have attracted much attention. This review focuses on alginate as a preferred polymer and presents recent advances in alginate-based polymers for probiotic delivery systems. We highlight several alginate-based delivery systems containing various types of probiotics and the physical and chemical modifications with chitosan, cellulose, starch, protein, fish gel, and many other materials to enhance their performance, of which the viability and protective mechanisms are discussed. Withal, various challenges in alginate-based polymers for probiotics delivery systems are traced out, and future directions, specifically on the use of nanomaterials as well as prebiotics, are delineated to further facilitate subsequent researchers in selecting more favorable materials and technology for probiotic delivery.

14 citations

Journal ArticleDOI
TL;DR: An amendment to this paper has been published and can be accessed via the original article.
Abstract: An amendment to this paper has been published and can be accessed via the original article.

4 citations

Journal ArticleDOI
01 Jun 2021
TL;DR: In this article, the effect of thirdhand cigarette smoke exposure and exposure age on the gut microbiome diversity was investigated in C57BL/6 mice, which were exposed to THS at human exposure relevant levels for three weeks during three different life stages: postnatal, pubescent and adult.
Abstract: The gut microbiome composition is influenced by many factors including environmental exposures. Here, we investigated the effect of thirdhand cigarette smoke (THS) and exposure age on gut microbiome diversity. C57BL/6 mice were exposed to THS at human exposure relevant levels for three weeks during three different life stages: postnatal (0-3 weeks of age), pubescent (4-7 weeks of age), and adult (9-12 weeks of age), respectively. Cecal microbiome profiles were assessed at 17 weeks of age by 16S rRNA gene sequencing. We found that age at THS exposure strongly influenced the cecal microbiome composition. Although postnatal THS exposure significantly influenced the microbial composition, pubescent and adulthood exposures only had minor effects. The microbiome of postnatally THS-exposed mice significantly increased several degradation pathways that regulate glycolysis and pyruvate decarboxylation, and significantly decreased coenzyme A biosynthesis and pyrimidine deoxyribonucleoside salvage. Our results indicate that mouse postnatal development is particularly susceptible to persistent THS exposure effects on the gut microbiome.

3 citations


Cited by
More filters
Journal ArticleDOI
TL;DR: In this article, the effects of the microbiota on host appetite and potential molecular mechanisms were discussed, and it was shown that the manipulation of the gut microbiota represents a clinical therapeutic potential for lessening the development and consequence of appetite-related disorders.
Abstract: Feelings of hunger and satiety are the key determinants for maintaining the life of humans and animals. Disturbed appetite control may disrupt the metabolic health of the host and cause various metabolic disorders. A variety of factors have been implicated in appetite control, including gut microbiota, which develop the intricate interactions to manipulate the metabolic requirements and hedonic feelings. Gut microbial metabolites and components act as appetite-related signaling molecules to regulate appetite-related hormone secretion and the immune system, or act directly on hypothalamic neurons. Herein, we summarize the effects of gut microbiota on host appetite and consider the potential molecular mechanisms. Furthermore, we propose that the manipulation of gut microbiota represents a clinical therapeutic potential for lessening the development and consequence of appetite-related disorders. Video abstract video file.(99M, mp4) Supplementary Information The online version contains supplementary material available at 10.1186/s40168-021-01093-y.

70 citations

Journal ArticleDOI
TL;DR: In this paper, the authors discuss the contribution of the intestinal microbiota to regulate systemic activities including gut-brain-immunity crosstalk, and discuss the companies worldwide translating the multi-therapeutic potential of human intestinal microbiota, based on multi-omics studies, as ready solutions for several metabolic and systemic disorders.
Abstract: The research on human gut microbiome, regarded as the black box of the human body, is still at the stage of infancy as the functional properties of the complex gut microbiome have not yet been understood. Ongoing metagenomic studies have deciphered that the predominant microbial communities belong to eubacterial phyla Firmicutes, Bacteroidetes, Proteobacteria, Fusobacteria, Cyanobacteria, Verrucomicrobia and archaebacterial phylum Euryarchaeota. The indigenous commensal microbial flora prevents opportunistic pathogenic infection and play undeniable roles in digestion, metabolite and signaling molecule production and controlling host's cellular health, immunity and neuropsychiatric behavior. Besides maintaining intestinal health via short-chain fatty acid (SCFA) production, gut microbes also aid in neuro-immuno-endocrine modulatory molecule production, immune cell differentiation and glucose and lipid metabolism. Interdependence of diet and intestinal microbial diversity suggests the effectiveness of pre- and pro-biotics in maintenance of gut and systemic health. Several companies worldwide have started potentially exploiting the microbial contribution to human health and have translated their use in disease management and therapeutic applications. The present review discusses the vast diversity of microorganisms playing intricate roles in human metabolism. The contribution of the intestinal microbiota to regulate systemic activities including gut-brain-immunity crosstalk has been focused. To the best of our knowledge, this review is the first of its kind to collate and discuss the companies worldwide translating the multi-therapeutic potential of human intestinal microbiota, based on the multi-omics studies, i.e. metagenomics and metabolomics, as ready solutions for several metabolic and systemic disorders.

37 citations

Journal ArticleDOI
TL;DR: A growing number of association studies exploring the gut microbiota's possible role in memory, learning, anxiety, stress, neurodevelopmental and neurodegenerative disorders have been conducted as mentioned in this paper .
Abstract: The gut and brain link via various metabolic and signalling pathways, each with the potential to influence mental, brain and cognitive health. Over the past decade, the involvement of the gut microbiota in gut-brain communication has become the focus of increased scientific interest, establishing the microbiota-gut-brain axis as a field of research. There is a growing number of association studies exploring the gut microbiota's possible role in memory, learning, anxiety, stress, neurodevelopmental and neurodegenerative disorders. Consequently, attention is now turning to how the microbiota can become the target of nutritional and therapeutic strategies for improved brain health and well-being. However, while such strategies that target the gut microbiota to influence brain health and function are currently under development with varying levels of success, still very little is yet known about the triggers and mechanisms underlying the gut microbiota's apparent influence on cognitive or brain function and most evidence comes from pre-clinical studies rather than well controlled clinical trials/investigations. Filling the knowledge gaps requires establishing a standardised methodology for human studies, including strong guidance for specific focus areas of the microbiota-gut-brain axis, the need for more extensive biological sample analyses, and identification of relevant biomarkers. Other urgent requirements are new advanced models for in vitro and in vivo studies of relevant mechanisms, and a greater focus on omics technologies with supporting bioinformatics resources (training, tools) to efficiently translate study findings, as well as the identification of relevant targets in study populations. The key to building a validated evidence base rely on increasing knowledge sharing and multi-disciplinary collaborations, along with continued public-private funding support. This will allow microbiota-gut-brain axis research to move to its next phase so we can identify realistic opportunities to modulate the microbiota for better brain health.

30 citations

Journal ArticleDOI
TL;DR: In this paper, the electrophysiological properties of hippocampal plasticity in adult germ-free mice and compare them to their conventionally raised counterparts were evaluated and it was shown that the absence of a microbiome alters integration of dendritic signalling in the CA1 region in mice.
Abstract: Germ-free rodents, raised in the absence of a measurable gut microbiome, have been a key model to study the microbiome-gut-brain axis. Germ-free mice exhibit marked behavioural and neurochemical differences to their conventionally raised counterparts. It is as yet unclear how these neurochemical differences lead to the behavioural differences. Here, we test the electrophysiological properties of hippocampal plasticity in adult germ-free mice and compare them to conventionally raised counterparts. Whilst basal synaptic efficacy and pre-synaptic short-term plasticity appear normal, we find a striking alteration of hippocampal long-term potentiation specifically in male germ-free slices. However, the spike output of these neurons remains normal along with altered input-output coupling, potentially indicating homeostatic compensatory mechanisms, or an altered excitation/inhibition balance. To our knowledge this is the first time the electrophysiological properties of the hippocampus have been assessed in a microbiome deficient animal. Our data indicate that the absence of a microbiome alters integration of dendritic signalling in the CA1 region in mice.

25 citations