Your browser doesn't support javascript.
loading
Gut dysbiosis induces the development of pre-eclampsia through bacterial translocation.
Chen, Xia; Li, Pan; Liu, Mian; Zheng, Huimin; He, Yan; Chen, Mu-Xuan; Tang, Wenli; Yue, Xiaojing; Huang, Yongxin; Zhuang, Lingling; Wang, Zhijian; Zhong, Mei; Ke, Guibao; Hu, Haoyue; Feng, Yinglin; Chen, Yun; Yu, Yanhong; Zhou, Hongwei; Huang, Liping.
Afiliação
  • Chen X; Department of Obstetrics and Gynecology, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China.
  • Li P; Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
  • Liu M; Department of Obstetrics and Gynecology, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China.
  • Zheng H; Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
  • He Y; Microbiome Research Centre, St George and Sutherland Clinical School, University of New South Wales, Sydney, New South Wales, Australia.
  • Chen MX; Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
  • Tang W; Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
  • Yue X; Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
  • Huang Y; Department of Obstetrics and Gynecology, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China.
  • Zhuang L; Department of Obstetrics and Gynecology, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China.
  • Wang Z; Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
  • Zhong M; Department of Obstetrics and Gynecology, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China.
  • Ke G; Department of Obstetrics and Gynecology, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China.
  • Hu H; Department of Internal Medicine, Affiliated Hospital, Chengdu University, Chengdu, Sichuan, China.
  • Feng Y; Department of Obstetrics and Gynecology, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China.
  • Chen Y; Department of Obstetrics and Gynecology, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China.
  • Yu Y; Department of Obstetrics and Gynecology, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China.
  • Zhou H; Department of Obstetrics and Gynecology, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China lphuang2006@126.com hzhou@smu.edu.cn yuyh1010@hotmail.com.
  • Huang L; Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China lphuang2006@126.com hzhou@smu.edu.cn yuyh1010@hotmail.com.
Gut ; 69(3): 513-522, 2020 03.
Article em En | MEDLINE | ID: mdl-31900289
ABSTRACT

OBJECTIVE:

Pre-eclampsia (PE) is one of the malignant metabolic diseases that complicate pregnancy. Gut dysbiosis has been identified for causing metabolic diseases, but the role of gut microbiome in the pathogenesis of PE remains unknown.

DESIGN:

We performed a case-control study to compare the faecal microbiome of PE and normotensive pregnant women by 16S ribosomal RNA (rRNA) sequencing. To address the causative relationship between gut dysbiosis and PE, we used faecal microbiota transplantation (FMT) in an antibiotic-treated mouse model. Finally, we determined the microbiome translocation and immune responses in human and mouse placental samples by 16S rRNA sequencing, quantitative PCR and in situ hybridisation.

RESULTS:

Patients with PE showed reduced bacterial diversity with obvious dysbiosis. Opportunistic pathogens, particularly Fusobacterium and Veillonella, were enriched, whereas beneficial bacteria, including Faecalibacterium and Akkermansia, were markedly depleted in the PE group. The abundances of these discriminative bacteria were correlated with blood pressure (BP), proteinuria, aminotransferase and creatinine levels. On successful colonisation, the gut microbiome from patients with PE triggered a dramatic, increased pregestational BP of recipient mice, which further increased after gestation. In addition, the PE-transplanted group showed increased proteinuria, embryonic resorption and lower fetal and placental weights. Their T regulatory/helper-17 balance in the small intestine and spleen was disturbed with more severe intestinal leakage. In the placenta of both patients with PE and PE-FMT mice, the total bacteria, Fusobacterium, and inflammatory cytokine levels were significantly increased.

CONCLUSIONS:

This study suggests that the gut microbiome of patients with PE is dysbiotic and contributes to disease pathogenesis.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Placenta / Pré-Eclâmpsia / Translocação Bacteriana / Disbiose / Microbioma Gastrointestinal Tipo de estudo: Observational_studies / Risk_factors_studies Limite: Animals / Female / Humans / Pregnancy Idioma: En Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Placenta / Pré-Eclâmpsia / Translocação Bacteriana / Disbiose / Microbioma Gastrointestinal Tipo de estudo: Observational_studies / Risk_factors_studies Limite: Animals / Female / Humans / Pregnancy Idioma: En Ano de publicação: 2020 Tipo de documento: Article