gut-lung axis

肠肺轴
  • 文章类型: Journal Article
    胃肠道含有由细菌组成的多种微生物组,真菌,病毒和古细菌。尽管这些微生物通常作为共生生物存在,现在已经确定,特定细菌或真菌物种的丰度更高,或者微生物组多样性的丧失会显著影响发育,疾病的进展和结果。研究主要集中在细菌的影响,然而,其他微生物的影响,比如真菌,在过去的几年里受到了越来越多的关注。真菌仅占总肠微生物种群的约0.1%。然而,关键真菌类群,如念珠菌,曲霉菌和Wallemia已被证明对健康和疾病产生重大影响。肠道真菌的组成已被证明会影响远端部位的免疫力,比如心脏,肺,大脑,胰腺,还有肝脏.在肺的情况下,这种现象被称为“肠-肺轴”。最近的研究已经开始探索和揭示在哮喘和肺癌等疾病中肠道真菌和肺部免疫之间的关系。病毒引起的肺部感染,细菌和真菌。在这篇综述中,我们将总结当前,快速增长,文献描述了肠道真菌对呼吸道疾病和感染的影响。
    The gastrointestinal tract contains a diverse microbiome consisting of bacteria, fungi, viruses and archaea. Although these microbes usually reside as commensal organisms, it is now well established that higher abundance of specific bacterial or fungal species, or loss of diversity in the microbiome can significantly affect development, progression and outcomes in disease. Studies have mainly focused on the effects of bacteria, however, the impact of other microbes, such as fungi, has received increased attention in the last few years. Fungi only represent around 0.1% of the total gut microbial population. However, key fungal taxa such as Candida, Aspergillus and Wallemia have been shown to significantly impact health and disease. The composition of the gut mycobiome has been shown to affect immunity at distal sites, such as the heart, lung, brain, pancreas, and liver. In the case of the lung this phenomenon is referred to as the \'gut-lung axis\'. Recent studies have begun to explore and unveil the relationship between gut fungi and lung immunity in diseases such as asthma and lung cancer, and lung infections caused by viruses, bacteria and fungi. In this review we will summarize the current, rapidly growing, literature describing the impact of the gut mycobiome on respiratory disease and infection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:肠-肺轴,对呼吸健康至关重要,在肺部和重症监护医学(PCCM)住院患者中没有充分探索。
    方法:检查来自三家医科大学附属医院的PCCM住院患者,我们对粪便样本进行了16S核糖体RNA测序(住院患者,n=374;健康对照,n=105)。我们进行了统计分析,以检查PCCM住院患者的肠道菌群组成,将其与健康对照进行比较。此外,我们探索了肠道菌群组成与各种临床因素之间的关联,包括年龄,白细胞计数,中性粒细胞计数,血小板计数,白蛋白水平,血红蛋白水平,住院时间,和医疗费用。
    结果:PCCM住院患者的肠道菌群多样性低于健康对照组。主坐标分析显示出明显的总体微生物群结构差异。四种肠型,包括住院患者独有的肠球菌科肠型,已确定。尽管在门一级没有发现任何区别,15个细菌家族表现出不同的丰度。具体来说,PCCM的住院人群显示出肠球菌科的丰度明显更高,乳酸杆菌科,丹毒科,梭菌科,和制革菌。使用随机森林分析,我们计算的受试者工作特征曲线下面积(AUC)为0.75(95%CI=0.69-0.80),用于区分健康个体和住院患者.分类器中保留的四个最丰富的属是Blautia,下颗粒,肠球菌,还有克雷伯菌.
    结论:PCCM住院患者肠道菌群失调的证据强调了肠-肺轴的意义,在呼吸健康研究中有希望的进一步途径。
    BACKGROUND: The gut-lung axis, pivotal for respiratory health, is inadequately explored in pulmonary and critical care medicine (PCCM) inpatients.
    METHODS: Examining PCCM inpatients from three medical university-affiliated hospitals, we conducted 16S ribosomal RNA sequencing on stool samples (inpatients, n = 374; healthy controls, n = 105). We conducted statistical analyses to examine the gut microbiota composition in PCCM inpatients, comparing it to that of healthy controls. Additionally, we explored the associations between gut microbiota composition and various clinical factors, including age, white blood cell count, neutrophil count, platelet count, albumin level, hemoglobin level, length of hospital stay, and medical costs.
    RESULTS: PCCM inpatients exhibited lower gut microbiota diversity than healthy controls. Principal Coordinates Analysis revealed marked overall microbiota structure differences. Four enterotypes, including the exclusive Enterococcaceae enterotype in inpatients, were identified. Although no distinctions were found at the phylum level, 15 bacterial families exhibited varying abundances. Specifically, the inpatient population from PCCM showed a significantly higher abundance of Enterococcaceae, Lactobacillaceae, Erysipelatoclostridiaceae, Clostridiaceae, and Tannerellaceae. Using random forest analyses, we calculated the areas under the receiver operating characteristic curves (AUCs) to be 0.75 (95% CIs 0.69-0.80) for distinguishing healthy individuals from inpatients. The four most abundant genera retained in the classifier were Blautia, Subdoligranulum, Enterococcus, and Klebsiella.
    CONCLUSIONS: Evidence of gut microbiota dysbiosis in PCCM inpatients underscores the gut-lung axis\'s significance, promising further avenues in respiratory health research.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    简介:流感病毒主要针对呼吸道,然而,在感染期间,呼吸系统和肠道系统都会受到损害。肺和肠损伤之间的联系仍不清楚。
    我们的实验采用16SrRNA技术和液相色谱-质谱(LC-MS)来检测流感病毒感染对小鼠粪便含量和代谢产物的影响。此外,通过HE染色研究流感病毒感染对肠道损伤的影响及其机制,蛋白质印迹,Q-PCR,和流式细胞术。
    我们的研究发现,流感病毒感染对肺部和肠道造成了重大损害,病毒只在肺部检测到。抗生素治疗加重了肺和肠损伤的严重程度。此外,感染后肺中Toll样受体7(TLR7)和干扰素-b(IFN-b)的mRNA水平显着增加。对肠道微生物群的分析显示,流感感染后成分发生了显着变化,包括增加的肠杆菌科和减少的乳杆菌科。相反,抗生素治疗减少了微生物多样性,特别是影响Firmicutes,变形杆菌,和拟杆菌。代谢组学显示由于流感感染和抗生素引起的氨基酸代谢途径的改变。吲哚胺2,3-双加氧酶1(IDO1)在结肠中的异常表达破坏了肠道中辅助T17细胞(Th17)和调节性T细胞(Treg细胞)之间的平衡。感染流感病毒并补充色氨酸和乳酸菌的小鼠显示出减少的肺和肠道损伤,肠杆菌在肠道中的水平降低,IDO1活性下降。
    总的来说,流感感染对肺和肠组织造成损害,肠道微生物群和代谢产物被破坏,影响Th17/Treg平衡。抗生素治疗加剧了这些影响。补充色氨酸和乳酸菌可改善肺部和肠道健康,强调对流感引起的肠道疾病中肺-肠连接的新认识。
    UNASSIGNED: Introduction: The influenza virus primarily targets the respiratory tract, yet both the respiratory and intestinal systems suffer damage during infection. The connection between lung and intestinal damage remains unclear.
    UNASSIGNED: Our experiment employs 16S rRNA technology and Liquid Chromatography-Mass Spectrometry (LC-MS) to detect the impact of influenza virus infection on the fecal content and metabolites in mice. Additionally, it investigates the effect of influenza virus infection on intestinal damage and its underlying mechanisms through HE staining, Western blot, Q-PCR, and flow cytometry.
    UNASSIGNED: Our study found that influenza virus infection caused significant damage to both the lungs and intestines, with the virus detected exclusively in the lungs. Antibiotic treatment worsened the severity of lung and intestinal damage. Moreover, mRNA levels of Toll-like receptor 7 (TLR7) and Interferon-b (IFN-b) significantly increased in the lungs post-infection. Analysis of intestinal microbiota revealed notable shifts in composition after influenza infection, including increased Enterobacteriaceae and decreased Lactobacillaceae. Conversely, antibiotic treatment reduced microbial diversity, notably affecting Firmicutes, Proteobacteria, and Bacteroidetes. Metabolomics showed altered amino acid metabolism pathways due to influenza infection and antibiotics. Abnormal expression of indoleamine 2,3-dioxygenase 1 (IDO1) in the colon disrupted the balance between helper T17 cells (Th17) and regulatory T cells (Treg cells) in the intestine. Mice infected with the influenza virus and supplemented with tryptophan and Lactobacillus showed reduced lung and intestinal damage, decreased Enterobacteriaceae levels in the intestine, and decreased IDO1 activity.
    UNASSIGNED: Overall, influenza infection caused damage to lung and intestinal tissues, disrupted intestinal microbiota and metabolites, and affected Th17/Treg balance. Antibiotic treatment exacerbated these effects. Supplementation with tryptophan and Lactobacillus improved lung and intestinal health, highlighting a new understanding of the lung-intestine connection in influenza-induced intestinal disease.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    临床前的证据已经确定了肺之间的双向相互作用,gut,gut和肠道微生物组。肺和肠之间有许多复杂的交流途径,这会影响彼此的平衡。一些由肠道微生物产生的代谢产物,肠道免疫细胞,免疫因子通过血液循环进入肺组织参与肺免疫功能。已经在啮齿动物模型和人类中发现了改变的肠-肺-微生物组相互作用,例如肺纤维化,慢性阻塞性肺疾病,肺癌,哮喘,等。新的证据表明,微生物疗法可以预防和治疗呼吸系统疾病,但尚不清楚这种关联是否与疾病的病理机制或因果关系简单相关。在这次审查中,我们总结了肠道菌群和肺之间复杂而关键的联系,以及肠道菌群对呼吸系统疾病的影响和机制,探讨益生元、粪菌移植等干预措施对呼吸系统疾病的作用。为大规模临床研究的合理设计提供参考,将微生物疗法直接应用于呼吸系统相关疾病可以降低疾病的发生率和严重程度以及伴随的并发症。
    Preclinical evidence has firmly established a bidirectional interaction among the lung, gut, and gut microbiome. There are many complex communication pathways between the lung and intestine, which affect each other\'s balance. Some metabolites produced by intestinal microorganisms, intestinal immune cells, and immune factors enter lung tissue through blood circulation and participate in lung immune function. Altered gut-lung-microbiome interactions have been identified in rodent models and humans of several lung diseases such as pulmonary fibrosis, chronic obstructive pulmonary disease, lung cancer, asthma, etc. Emerging evidence suggests that microbial therapies can prevent and treat respiratory diseases, but it is unclear whether this association is a simple correlation with the pathological mechanisms of the disease or the result of causation. In this review, we summarize the complex and critical link between the gut microbiota and the lung, as well as the influence and mechanism of the gut microbiota on respiratory diseases, and discuss the role of interventions such as prebiotics and fecal bacteria transplantation on respiratory diseases. To provide a reference for the rational design of large-scale clinical studies, the direct application of microbial therapy to respiratory-related diseases can reduce the incidence and severity of diseases and accompanying complications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    生命早期使用抗生素会破坏微生物定植,并增加发生过敏和哮喘的风险。我们报告说,小鼠在生命早期给予抗生素(EL-Abx),但不是在成年后,更容易受到屋尘螨(HDM)引起的过敏性气道炎症。即使在肠道微生物组正常化后,这种易感性仍得以维持。EL-Abx降低吲哚-3-丙酸(IPA)的全身水平,诱导细胞应激的长期变化,新陈代谢,和肺上皮的线粒体呼吸。IPA减少线粒体呼吸和超氧化物产生并改变趋化因子和细胞因子产生。因此,早期补充IPA可保护EL-Abx小鼠在成年期抵抗HDM引起的过敏性气道炎症。这些结果揭示了EL-Abx可以使肺易患过敏性气道炎症的机制,并强调了一种可能的预防方法来减轻EL-Abx的有害后果。
    Antibiotic use in early life disrupts microbial colonization and increases the risk of developing allergies and asthma. We report that mice given antibiotics in early life (EL-Abx), but not in adulthood, were more susceptible to house dust mite (HDM)-induced allergic airway inflammation. This susceptibility was maintained even after normalization of the gut microbiome. EL-Abx decreased systemic levels of indole-3-propionic acid (IPA), which induced long-term changes to cellular stress, metabolism, and mitochondrial respiration in the lung epithelium. IPA reduced mitochondrial respiration and superoxide production and altered chemokine and cytokine production. Consequently, early-life IPA supplementation protected EL-Abx mice against exacerbated HDM-induced allergic airway inflammation in adulthood. These results reveal a mechanism through which EL-Abx can predispose the lung to allergic airway inflammation and highlight a possible preventative approach to mitigate the detrimental consequences of EL-Abx.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    肺中性粒细胞增多症是许多气道疾病的标志,包括慢性阻塞性肺疾病(COPD),中性粒细胞性哮喘,急性肺损伤(ALI),急性呼吸窘迫综合征(ARDS)和COVID-19。本研究的目的是调查饮食干预对肺中性粒细胞增多的影响。
    雄性BALB/cByJ小鼠接受7个鼻内剂量的媒介物或脂多糖(LPS)。为了研究营养干预的效果,他们接受了16剂胃内剂量的媒介物(PBS)或以下补充剂(1)益生菌短双歧杆菌(B.breve)M16-V;(2)短链半乳寡糖的益生元纤维混合物,长链低聚果糖,和9:1:2比例的低粘度果胶(scGOS/lcFOS/lvPectin);和(3)合生元组合短芽孢杆菌M16-V和scGOS/lcFOS/lvPectin。肺健康参数包括肺功能,肺形态和肺部炎症。全身免疫调节的参数包括粪便短链脂肪酸和调节性T细胞的水平。
    合生元补充剂可防止LPS诱导的肺功能下降(基线p=0.0002时,肺阻力提高了35%,峰值时提高了25%,p=0.0002),提供了肺嗜中性粒细胞的显着缓解(减少了40.7%的嗜中性粒细胞,p<0.01),并将肺中性粒细胞与淋巴细胞的比率(NLR)提高了55.3%(p=0.0033)。在该特定实验中,补充剂不影响肺形态。与接受PBS的小鼠相比,应用于上气道的LPS诱导较少的粪便SCFA产生。在所有未攻击的小鼠中,在第-5天和第16天之间乙酸的产生增加(PBS-PBSp=0.0003;PBS-Prop<0.0001;PBS-Pre,p=0.0045;PBS-Syn,p=0.0005),仅在接受短双歧杆菌M16-V和GOS:FOS:lvPectin合生元混合物的小鼠中观察到LPS攻击(p=0.0003)。发现丁酸和肺功能参数之间存在中度相关性,发现乙酸之间存在弱相关性,丁酸和丙酸浓度和NLR。
    本研究提示了肺嗜中性粒细胞增多症的小鼠模型中的双向肠道肺串扰。中性粒细胞性肺部炎症与粪便SCFA水平减弱共存。短双歧杆菌M16-V和GOS:FOS:lvPectin的合生元混合物对与SCFA水平增强相关的肺健康的有益效果。
    UNASSIGNED: Pulmonary neutrophilia is a hallmark of numerous airway diseases including Chronic Obstructive Pulmonary Disease (COPD), Neutrophilic asthma, Acute Lung Injury (ALI), Acute Respiratory Distress Syndrome (ARDS) and COVID-19. The aim of the current study was to investigate the effect of dietary interventions on lung health in context of pulmonary neutrophilia.
    UNASSIGNED: Male BALB/cByJ mice received 7 intra-nasal doses of either a vehicle or lipopolysaccharides (LPS). To study the effect of nutritional interventions they received 16 intra-gastric doses of either a vehicle (PBS) or the following supplements (1) probiotic Bifidobacterium breve (B. breve) M16-V; (2) a prebiotic fiber mixture of short-chain galacto-oligosaccharides, long-chain fructo-oligosaccharides, and low-viscosity pectin in a 9:1:2 ratio (scGOS/lcFOS/lvPectin); and (3) A synbiotic combination B. breve M16-V and scGOS/lcFOS/lvPectin. Parameters for lung health included lung function, lung morphology and lung inflammation. Parameters for systemic immunomodulation included levels of fecal short chain fatty acids and regulatory T cells.
    UNASSIGNED: The synbiotic supplement protected against the LPS induced decline in lung function (35% improved lung resistance at baseline p = 0.0002 and 25% at peak challenge, p = 0.0002), provided a significant relief from pulmonary neutrophilia (40.7% less neutrophils, p < 0.01) and improved the pulmonary neutrophil-to-lymphocyte ratio (NLR) by 55.3% (p = 0.0033). Supplements did not impact lung morphology in this specific experiment. LPS applied to the upper airways induced less fecal SCFAs production compared to mice that received PBS. The production of acetic acid between day -5 and day 16 was increased in all unchallenged mice (PBS-PBS p = 0.0003; PBS-Pro p < 0.0001; PBS-Pre, p = 0.0045; PBS-Syn, p = 0.0005) which upon LPS challenge was only observed in mice that received the synbiotic mixture of B. breve M16-V and GOS:FOS:lvPectin (p = 0.0003). A moderate correlation was found for butyric acid and lung function parameters and a weak correlation was found between acetic acid, butyric acid and propionic acid concentrations and NLR.
    UNASSIGNED: This study suggests bidirectional gut lung cross-talk in a mouse model for pulmonary neutrophilia. Neutrophilic lung inflammation coexisted with attenuated levels of fecal SCFA. The beneficial effects of the synbiotic mixture of B. breve M16-V and GOS:FOS:lvPectin on lung health associated with enhanced levels of SCFAs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    最近的实验和流行病学研究强调了肠道微生物群和肺部之间的重要相互作用,一种被称为“肠-肺轴”的相互作用。在肠道微生物代谢产物的鉴定后,该轴的意义已被进一步阐明。例如短链脂肪酸(SCFA),作为设定免疫系统基调的关键媒介。通过肠-肺轴,肠道微生物群及其代谢产物,或者过敏原,直接或间接参与肺部疾病的免疫调节,从而增加对过敏性气道疾病如哮喘的易感性。哮喘是环境因素和遗传易感性之间相互作用的复杂结果。肠-肺轴的概念可能为哮喘的预防和治疗提供新的靶点。本文概述了哮喘与呼吸道微生物组之间的关系。肠道微生物组,和环境微生物组。它还讨论了微生物学的当前进展和应用,为哮喘等慢性呼吸系统疾病的临床管理提供了新的观点和策略。
    Recent experimental and epidemiological studies underscore the vital interaction between the intestinal microbiota and the lungs, an interplay known as the \"gut-lung axis\". The significance of this axis has been further illuminated following the identification of intestinal microbial metabolites, such as short-chain fatty acids (SCFA), as key mediators in setting the tone of the immune system. Through the gut-lung axis, the gut microbiota and its metabolites, or allergens, are directly or indirectly involved in the immunomodulation of pulmonary diseases, thereby increasing susceptibility to allergic airway diseases such as asthma. Asthma is a complex outcome of the interplay between environmental factors and genetic predispositions. The concept of the gut-lung axis may offer new targets for the prevention and treatment of asthma. This review outlines the relationships between asthma and the respiratory microbiome, gut microbiome, and environmental microbiome. It also discusses the current advancements and applications of microbiomics, offering novel perspectives and strategies for the clinical management of chronic respiratory diseases like asthma.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:肠道菌群(GM)被认为是胃肠道症状的重要调节因子,这种症状通常与呼吸道甲型流感病毒(IAV)感染一起发生,提示肠-肺轴参与宿主对IAV的反应。IAV主要破坏气道上皮紧密连接(TJ),并因此引起急性呼吸道疾病综合征。众所周知,转基因及其代谢产生抗流感作用,但它们在IAV诱导的气道上皮完整性中的作用尚不清楚.
    方法:建立小鼠IAV感染模型。使用16SrRNA基因测序分析GM,测量短链脂肪酸(SCFA)水平。进行了GM消耗和粪便微生物群移植(FMT)以验证GM在IAV感染中的作用。进行了配对喂养实验,以揭示IAV诱导的转基因生态失调是否归因于食物摄入受损。此外,在存在或不存在乙酸盐的情况下,将人支气管上皮(HBE)细胞与IAV共培养。通过细胞旁通透性和跨上皮电子电阻(TEER)分析TJs功能。在用G蛋白偶联受体43(GPR43)短发夹RNA(shRNA)转染的HBE细胞中评估了乙酸盐如何影响TJ完整性的机制。
    结果:感染IAV的小鼠表现出较低的产乙酸细菌相对丰度(拟杆菌,双歧杆菌,和Akkermansia)和肠道和血清中的乙酸盐水平降低。这些变化部分是由食物消耗减少(由于厌食症)引起的。GM耗竭加剧,FMT恢复了IAV诱导的肺部炎症损伤。IAV感染抑制了TJs的表达(occludin,ZO-1)导致气道上皮屏障功能被破坏,如TEER降低和通透性增加所证明。醋酸预处理激活GPR43,部分恢复IAV诱导的气道上皮屏障功能,和降低炎症细胞因子水平(TNF-α,IL-6和IL-1β)。在用GPR43shRNA转染的HBE细胞中不存在乙酸盐的这种保护作用。醋酸盐和GPR43以AMP激活的蛋白激酶(AMPK)依赖性方式改善了TJ。
    结论:总的来说,我们的结果表明,在IAV诱导的肺损伤中,GM通过调节GPR43-AMPK信号通路保护气道TJs.因此,改善GM菌群失调可能是IAV感染患者的潜在治疗目标。
    BACKGROUND: Gut microbiota (GM) have been implicated as important regulators of gastrointestinal symptom which is commonly occurred along with respiratory influenza A virus (IAV) infection, suggesting the involvement of the gut-to-lung axis in a host\'s response to IAV. IAV primarily destroys airway epithelium tight junctions (TJs) and consequently causes acute respiratory disease syndrome. It is known that GM and their metabolism produce an anti-influenza effect, but their role in IAV-induced airway epithelial integrity remains unknown.
    METHODS: A mouse model of IAV infection was established. GM were analyzed using 16S rRNA gene sequencing, and short-chain fatty acids (SCFAs) levels were measured. GM depletion and fecal microbiota transplantation (FMT) were conducted to validate the role of GM in IAV infection. A pair-feeding experiment was conducted to reveal whether IAV-induced GM dysbiosis is attributed to impaired food intake. Furthermore, human bronchial epithelial (HBE) cells were cocultured with IAV in the presence or absence of acetate. TJs function was analyzed by paracellular permeability and transepithelial electronic resistance (TEER). The mechanism of how acetate affects TJs integrity was evaluated in HBE cells transfected with G protein-coupled receptor 43 (GPR43) short hairpin RNA (shRNA).
    RESULTS: IAV-infected mice exhibited lower relative abundance of acetate-producing bacteria (Bacteroides, Bifidobacterium, and Akkermansia) and decreased acetate levels in gut and serum. These changes were partly caused by a decrease in food consumption (due to anorexia). GM depletion exacerbated and FMT restored IAV-induced lung inflammatory injury. IAV infection suppressed expressions of TJs (occludin, ZO-1) leading to disrupted airway epithelial barrier function as evidenced by decreased TEER and increased permeability. Acetate pretreatment activated GPR43, partially restored IAV-induced airway epithelial barrier function, and reduced inflammatory cytokines levels (TNF-α, IL-6, and IL-1β). Such protective effects of acetate were absent in HBE cells transfected with GPR43 shRNA. Acetate and GPR43 improved TJs in an AMP-activated protein kinase (AMPK)-dependent manner.
    CONCLUSIONS: Collectively, our results demonstrated that GM protected airway TJs by modulating GPR43-AMPK signaling in IAV-induced lung injury. Therefore, improving GM dysbiosis may be a potential therapeutic target for patients with IAV infection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号