首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
哺乳动物的肠道内栖息着庞大复杂的微生物群体,其微生物群体与宿主的消化吸收、物质的营养代谢和免疫功能密切相关,是影响机体健康的重要因素之一。随着分子生物学技术在肠道微生物领域的应用,特别是新一代测序技术的快速发展,使得人们对复杂的肠道微生物的研究更加深入。基于宏基因组学技术不仅能够研究肠道微生物组的多样性、揭示消化道微生物对宿主生理代谢的影响,还能进一步深入挖掘新的功能基因,并揭示宿主基因与微生物组间的互作关系和共同进化。作者综述了宏基因组学技术在哺乳动物肠道微生物中的主要应用和存在的不足,并展望了其在肠道微生物研究中的广阔应用前景,从而加深人们对肠道微生物影响宿主肠道健康作用的认识。  相似文献   

2.
3.
The gut microbiome has long been known to play fundamentally important roles in the animal health and the well-being of its host. As such, the establishment and maintenance of a beneficial gut microbiota early in life is crucial in pigs, since early gut colonizers are pivotal in the establishment of permanent microbial community structures affecting the health and growth performance of pigs later in life. Emphasizing this importance of early gut colonizers, it is critical to understand the factors impacting the establishment of the piglet gut microbiome at weaning. Factors include, among others, diet, in-feed antibiotics, probiotics and prebiotic administration. The impact of these factors on establishment of the gut microbiome of piglets at weaning includes effects on piglet gut microbial diversity, structure, and succession. In this review, we thoroughly reviewed the most recent findings on the piglet gut microbiome shifts as influenced by weaning, and how these microbiome changes brought about by various factors that have been shown to affect the development of microbiota in piglets. This review will provide a general overview of recent studies that can help to facilitate the design of new strategies to modulate the gut microbiome in order to enhance gastrointestinal health, growth performance and well-being of piglets.  相似文献   

4.
Information on the gut microbiome composition of different mammals could provide novel insights into the evolution of mammals and succession of microbial communities in different hosts. However, there is limited information on the gut microbiome composition of marine mammals, especially cetaceans because of sampling constraints. In this study, we investigated the diversity and composition of microbial communities in the stomach, midgut, and hindgut of 2 stranded short-finned pilot whales (Globicephala macrorhynchus) and hindgut of a stranded pygmy sperm whale (Kogia breviceps) by using 16S rRNA gene amplicon sequencing technology. On the basis of the 50 most abundant operational taxonomic units, principal coordinate analysis, and non-metric multidimensional scaling analysis, we confirmed that the gut microbial communities of the 3 whales were different. Our results revealed that the gut microbiome of 1 stranded short-finned pilot whale GM16 was dominated by Firmicutes (mainly Clostridium) and Fusobacteria; whereas that of the other pilot whale GM19 was composed of Gammaproteobacteria and Bacteroidetes (mainly Vibrio and Bacteroides, respectively), probably caused by intestinal disease and antibiotic treatment. The gut microbiome of the pygmy sperm whale was dominated by Firmicutes and Bacteroidetes. Moreover, different gastrointestinal tract regions harbored different microbial community structures. To our knowledge, this is the first report of the gut microbiome of short-finned pilot whales, and our findings will expand our current knowledge on microbial diversity and composition in the gastrointestinal tract of cetaceans.  相似文献   

5.
Gut microbiota is generally recognized to play a crucial role in maintaining host health and metabolism. The correlation among gut microbiota, glycolipid metabolism, and metabolic diseases has been well reviewed in humans. However, the interplay between gut microbiota and host metabolism in swine remains incompletely understood. Given the limitation in conducting human experiments and the high similarity between swine and humans in terms of anatomy, physiology, polyphagy, habits, and metabolism and in terms of the composition of gut microbiota, there is a pressing need to summarize the knowledge gained regarding swine gut microbiota, its interplay with host metabolism, and the underlying mechanisms. This review aimed to outline the bidirectional regulation between gut microbiota and nutrient metabolism in swine and to emphasize the action mechanisms underlying the complex microbiome–host crosstalk via the gut microbiota–gut–brain axis. Moreover, it highlights the new advances in knowledge of the diurnal rhythmicity of gut microbiota. A better understanding of these aspects can not only shed light on healthy and efficient pork production but also promote our knowledge on the associations between gut microbiota and the microbiome–host crosstalk mechanism. More importantly, knowledge on microbiota, host health and metabolism facilitates the development of a precise intervention therapy targeting the gut microbiota.  相似文献   

6.
Cancer is a substantial global health problem both in humans and animals with a consistent increase in mortality and incidence rate. The commensal microbiota has been involved in the regulation of several physiological and pathological processes, both within the gastrointestinal system and at distant tissue locations. Cancer is not an exception, and different aspects of the microbiome have been described to have anti- or pro-tumour effects. Using new techniques, for example high-throughput DNA sequencing, microbial populations of the human body have been largely described and, in the last years, studies more focused on companions' animals have emerged. In general, the recent investigations of faecal microbial phylogeny and functional capacity of the canine and feline gut have shown similarities with human gut. In this translational study we will review and summarize the relation between the microbiota and cancer, in humans and companion animals, and compare their resemblance in the type of neoplasms already studied in veterinary medicine: multicentric and intestinal lymphoma, colorectal tumours, nasal neoplasia and mast cell tumours. In the context of One Health, microbiota and microbiome integrative studies may contribute to the understanding of the tumourigenesis process, besides offering an opportunity to develop new diagnostics and therapeutic biomarkers both for veterinary and human oncology.  相似文献   

7.
Metals such as iron, manganese, copper, and zinc are recognized as essential trace elements. These trace metals play critical roles in development, growth, and metabolism, participating in various metabolic processes by acting as cofactors of enzymes or providing structural support to proteins. Deficiency or toxicity of these metals can impact human and animal health, giving rise to a number of metabolic and neurological disorders. Proper breakdown, absorption, and elimination of these trace metals is a tightly regulated process that requires crosstalk between the host and these micronutrients. The gut is a complex system that serves as the interface between these components, but other factors that contribute to this delicate interaction are not well understood. The gut is home to trillions of microorganisms and microbial genes (the gut microbiome) that can regulate the metabolism and transport of micronutrients and contribute to the bioavailability of trace metals through their assimilation from food sources or by competing with the host. Furthermore, deficiency or toxicity of these metals can modulate the gut microenvironment, including microbiota, nutrient availability, stress, and immunity. Thus, understanding the role of the gut microbiota in the metabolism of manganese, iron, copper, and zinc, as well as in heavy metal deficiencies and toxicities, and vice versa, may provide insight into developing improved or alternative therapeutic strategies to address emerging health concerns. This review describes the current understanding of how the gut microbiome and trace metals interact and affect host health, particularly in pigs.  相似文献   

8.
The gastrointestinal tract contains within it a microenvironment of bacteria that influences the host animal in many ways. The microflora can metabolize several nutrients that the host cannot digest and converts these to end products (such as short-chain fatty acids), a process that has a direct impact on digestive physiology. The microbiota directs the assembly of the gut-associated lymphoid tissue, helps educate the immune system, affects the integrity of the intestinal mucosal barrier, modulates proliferation and differentiation of its epithelial lineages, regulates angiogenesis, modifies the activity of the enteric nervous system, and plays a key role in extracting and processing nutrients consumed in the diet. Despite these important effects, the mechanisms by which the gut microbial community influences host biology remain almost entirely unknown. Recent molecular-based investigations have confirmed the species diversity and metabolic complexity of gut microflora, although there is much work to be done to understand how they relate to each other as well as the host animal. It is almost a century ago that Eli Metchnikoff proposed the revolutionary idea to consume viable bacteria to promote health. Since that time, the area known as probiotics has made dramatic progress, particularly during the past 2 decades. The last 20 yr have also seen the emergence of a new, related area of study—prebiotics. Use of these 2 ideas—providing live nonpathogenic bacteria as well as substrates for their growth—have potential to help optimize the health of animals by manipulating the gastrointestinal tract in positive ways.  相似文献   

9.
Colonization and development of the gut microbiome is a crucial consideration for optimizing the health and performance of livestock animals. This is mainly attributed to the fact that dietary and management practices greatly influence the gut microbiota, subsequently leading to changes in nutrient utilization and immune response. A favorable microbiome can be implanted through dietary or management interventions of livestock animals, especially during early life. In this review, we explore all the possible factors (for example gestation, colostrum, and milk feeding, drinking water, starter feed, inoculation from healthy animals, prebiotics/probiotics, weaning time, essential oil and transgenesis), which can influence rumen microbiome colonization and development. We discuss the advantages and disadvantages of potential strategies used to manipulate gut development and microbial colonization to improve the production and health of newborn calves at an early age when they are most susceptible to enteric disease. Moreover, we provide insights into possible interventions and their potential effects on rumen development and microbiota establishment. Prospects of latest techniques like transgenesis and host genetics have also been discussed regarding their potential role in modulation of rumen microbiome and subsequent effects on gut development and performance in neonatal ruminants.  相似文献   

10.
The intestinal tract is a host to 100 trillion of microbes that have co-evolved with mammals over the millennia. These commensal organisms are critical to the host survival. The roles that symbiotic microorganisms play in the digestion, absorption, and metabolism of nutrients have been clearly demonstrated. Additionally, commensals are indispensable in regulating host immunity. This is evidenced by the poorly developed gut immune system of germ-free mice, which can be corrected by transplantation of specific commensal bacteria. Recent advances in our understanding of the mechanism of host–microbial interaction have provided the basis for this interaction. This paper reviews some of these key studies, with a specific focus on the effect of the microbiome on the immune organ development, nonspecific immunity, specific immunity, and inflammation.  相似文献   

11.
Studies on stress and its impacts on animals are very important in many fields of science, including animal science, because various stresses influence animal production and animal welfare. In particular, the social stresses within animal groups have profound impact on animals, with the potential to induce abnormal behaviors and health problems. In humans, social stress induces several health problems, including psychiatric disorders. In animal stress models, social defeat models are well characterized and used in various research fields, particularly in studies concerning mental disorders. Recently, we have focused on behavior, nutrition and metabolism in rodent models of social defeat to elucidate how social stresses affect animals. In this review, recent significant progress in studies related to animal social defeat models are described. In the field of animal science, these stress models may contribute to advances in the development of functional foods and in the management of animal welfare.  相似文献   

12.
Rumen microbiota has a close and intensive interaction with the ruminants. Microbiota residing in the rumen digests and ferments plant organic matters into nutrients that are subsequently utilized by the host, making ruminants a unique group of animals that can convert plant materials indigestible by humans into high-quality animal protein as meat and milk. Many studies using meta-omics technologies have demonstrated the relationships between rumen microbiome and animal phenotypes associated with nutrient metabolism. Recently, the causality and physiological mechanisms underpinning the host–microbiota interactions have attracted tremendous research interest among researchers. This review discusses the host–microbiota interactions and the factors affecting these interactions in ruminants and provides a summary of the advances in research on animal husbandry. Understanding the microbiota composition, the functions of key bacteria, and the host–microbiota interaction is crucial for the development of knowledge-based strategies to enhance animal productivity and host health.  相似文献   

13.
The priorities of public health and agricultural sciences intersect through a shared objective to foster better human health. Enhancements in food quality and reductions in the environmental effects of modern agriculture represent 2 distinct paths through which animal sciences can contribute to the cause of public health. Recent developments in the study of human-associated microbial communities (microbiotas), notably in association with disease, indicate that better understanding of the microbial ecology of livestock can contribute to achieving the goals of better foods and a cleaner environment. Culture-independent microbiological technologies now permit comprehensive study of complex microbial communities in their natural environments. Microbiotas associated with both humans and animals provide myriad beneficial services to their hosts that, if lost or diminished, could compromise host health. Dysfunctional microbial communities have been noted in several human conditions, including inflammatory bowel disease, obesity, and antibiotic-associated diarrhea. Examination of the mechanisms by which the human microbiota influences health and disease susceptibility can inform similar studies of host-microbe function in the animal sciences. Insights gained from human studies indicate strategies to raise not only healthier livestock, through selective manipulation of microbial communities, but also healthier humans.  相似文献   

14.
Recent molecular studies have revealed complex bacterial, fungal, archaeal, and viral communities in the gastrointestinal tract of dogs and cats. More than 10 bacterial phyla have been identified, with Firmicutes, Bacteroidetes, Proteobacteria, Fusobacteria, and Actinobacteria constituting more than 99% of all gut microbiota. Microbes act as a defending barrier against invading pathogens, aid in digestion, provide nutritional support for enterocytes, and play a crucial role in the development of the immune system. Of significance for gastrointestinal health is their ability to ferment dietary substrates into short-chain fatty acids, predominantly to acetate, propionate, and butyrate. However, microbes can have also a detrimental effect on host health. Specific pathogens (e.g., Salmonella, Campylobacter jejuni, and enterotoxigenic Clostridium perfringens) have been implicated in acute and chronic gastrointestinal disease. Compositional changes in the small intestinal microbiota, potentially leading to changes in intestinal permeability and digestive function, have been suggested in canine small intestinal dysbiosis or antibiotic-responsive diarrhea. There is mounting evidence that microbes play an important role in the pathogenesis of canine and feline inflammatory bowel disease (IBD). Current theories for the development of IBD favor a combination of environmental factors, the intestinal microbiota, and a genetic susceptibility of the host. Recent studies have revealed a genetic susceptibility for defective bacterial clearance in Boxer dogs with granulomatous colitis. Differential expression of pathogen recognition receptors (i.e., Toll-like receptors) were identified in dogs with chronic enteropathies. Similarly to humans, a microbial dysbiosis has been identified in feline and canine IBD. Commonly observed microbial changes are increased Proteobacteria (i.e., Escherichia coli) with concurrent decreases in Firmicutes, especially a reduced diversity in Clostridium clusters XIVa and IV (i.e., Lachnospiraceae, Ruminococcaceae, Faecalibacterium spp.). This would indicate that these bacterial groups, important short-chain fatty acid producers, may play an important role in promoting intestinal health.  相似文献   

15.
宏基因组学揭示瘤胃微生物多样性及功能   总被引:1,自引:0,他引:1  
反刍动物瘤胃内栖息着庞大和复杂的微生物群体,这些微生物与宿主的消化吸收、营养代谢和免疫功能息息相关,宿主及其微生物共同组成了一个"超级生物体"。由于绝大部分瘤胃微生物不可培养,因此以厌氧培养为基础的传统研究方法存在明显的弊端。宏基因组学通过高通量的测序方法,能够全面展示微生物多样性,准确发现新的功能基因。此外,宏基因组学揭示了宿主基因和微生物组之间的互作关系。随着组学技术的不断发展,宏基因组学在瘤胃微生物组研究方面具有广阔的应用前景。  相似文献   

16.
Gastrointestinal microbiota play a crucial role in nutrient digestion, maintaining animal health and welfare. Various factors may affect microbial balance often leading to disturbances that may result in debilitating conditions such as colic and laminitis. The invention of next-generation sequencing technologies and bioinformatics has provided valuable information on the effects of factors influencing equine gut microbiota. Among those factors are nutrition and management (e.g., diet, supplements, exercise), medical substances (e.g., antimicrobials, anthelmintics, anesthetics), animal-related factors (breed and age), various pathological conditions (colitis, diarrhea, colic, laminitis, equine gastric ulcer syndrome), as well as stress-related factors (transportation and weaning). The aim of this review is to assimilate current knowledge on equine microbiome studies, focusing on the effect of factors influencing equine gastrointestinal microbiota. Decrease in microbial diversity and richness leading to decrease in stability; decrease in Lachnospiraceae and Ruminococcaceae family members, which contribute to gut homeostasis; increase in Lactobacillus and Streptococcus; decrease in lactic acid utilizing bacteria; decrease in butyrate-producing bacteria that have anti-inflammatory properties may all be considered as a negative change in equine gut microbiota. Shifts in Firmicutes and Bacteroidetes have often been observed in the literature in response to certain treatments or when describing healthy and unhealthy animals; however, these shifts are inconsistent. It is time to move forward and use the knowledge now acquired to start manipulating the microbiota of horses.  相似文献   

17.
To achieve further reductions in foodborne illness levels in humans, effective pre-harvest interventions are needed. The health status of food animals that are destined to enter the human food supply chain may be an important, although often overlooked, factor in predicting the risk of human foodborne infections. The health status of food animals can potentially influence foodborne pathogen levels in three ways. First, diseased animals may shed higher levels of foodborne pathogens. Second, animals that require further handling in the processing plant to remove affected parts may lead to increased microbial contamination and cross-contamination. Finally, certain animal illnesses may lead to a higher probability of mistakes in the processing plant, such as gastrointestinal ruptures, which would lead to increased microbial contamination and cross-contamination. Consequently, interventions that reduce the incidence of food animal illnesses might also help reduce bacterial contamination on meat, thereby reducing human illness. Some of these interventions, however, might also present a risk to human health. For example, the use of antibiotics in food animals can reduce rates of animal illness but can also select for antibiotic-resistant bacteria which can threaten human treatment options. In this study, we present a mathematical model to evaluate human health risks from foodborne pathogens associated with changes in animal illness. The model is designed so that potential human health risks and benefits from interventions such as the continued use of antibiotics in animal agriculture can be evaluated simultaneously. We applied the model to a hypothetical example of Campylobacter from chicken. In general, the model suggests that very minor perturbations in microbial loads on meat products could have relatively large impacts on human health, and consequently, small improvements in food animal health might result in significant reductions in human illness.  相似文献   

18.
肠道微生物被称为动物的“隐藏免疫器官”,不仅能参与宿主代谢还能影响宿主的免疫系统,对维持机体健康至关重要。作者主要介绍了培养组学的发展历程及其对动物肠道微生物研究的重要意义、传统微生物培养方法和分子生物学方法在研究微生物时各自的优、缺点。培养组学是基于传统微生物培养方法同时采用多种培养条件进行微生物培养,再辅以基质辅助激光解吸电离飞行时间质谱(MALDI-TOF MS)和16S rRNA基因测序技术建立的一种新型微生物分离、鉴定方法,该方法将传统微生物培养技术与分子生物学技术的优点融为一体。该方法在挖掘“新微生物”的研究中,具有发现、找到并获得的优势;在微生物的研究中可定制分离目标菌株进行验证,并能通过丰富注释清楚地了解肠道微生物组。此外,分析了培养组学分别在家禽肠道、猪肠道、反刍动物肠道等动物肠道的研究应用现状,提出了环境条件对肠道微生物的影响,如人类接触对肠道菌群的影响、同物种不同性别肠道菌群的差异,以期为培养组学在动物肠道微生物的研究运用中提供参考。  相似文献   

19.
微生物饲料添加剂的主要功能及其研究进展   总被引:2,自引:0,他引:2  
益生菌是当摄入量足够时能对机体产生有益作用的活性微生物.人们常把应用于畜牧业生产上的益生菌称为微生物饲料添加剂.以往研究表明,微生物饲料添加剂具有维护动物肠道健康、缓解不良应激、改善畜舍环境、调节机体脂肪代谢和改善畜产品品质的功能.还有研究者认为,微生物饲料添加剂具有替代抗生素功能的作用.本文旨在就微生物饲料添加剂的主要功能及其研究进展进行综述,为其今后在畜牧业生产上的科学应用及相关研究提供参考.  相似文献   

20.
Polyphenols are a class of non-essential phytonutrients, which are abundant in fruits and vegetables. Dietary polyphenols or foods rich in polyphenols are widely recommended for metabolic health. Indeed, polyphenols (i.e., catechins, resveratrol, and curcumin) are increasingly recognized as a regulator of lipid metabolism in host. The mechanisms, at least in part, may be highly associated with gut microbiome. This review mainly discussed the beneficial effects of dietary polyphenols on lipid metabolism. The potential mechanisms of gut microbiome are focused on the effect of dietary polyphenols on gut microbiota compositions and how gut microbiota affect polyphenol metabolism. Together, dietary polyphenols may be a useful nutritional strategy for manipulation of lipid metabolism or obesity care.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号