Your browser doesn't support javascript.
loading
Suppression of Endothelial AGO1 Promotes Adipose Tissue Browning and Improves Metabolic Dysfunction.
Tang, Xiaofang; Miao, Yifei; Luo, Yingjun; Sriram, Kiran; Qi, Zhijie; Lin, Feng-Mao; Gu, Yusu; Lai, Chih-Hung; Hsu, Chien-Yi; Peterson, Kirk L; Van Keuren-Jensen, Kendall; Fueger, Patrick T; Yeo, Gene W; Natarajan, Rama; Zhong, Sheng; Chen, Zhen Bouman.
Affiliation
  • Tang X; Department of Diabetes Complications and Metabolism (X.T., Y.M., Y.L., K.S., F.L., C.H.L., R.N., Z.C.), City of Hope, Duarte, CA.
  • Miao Y; Department of Diabetes Complications and Metabolism (X.T., Y.M., Y.L., K.S., F.L., C.H.L., R.N., Z.C.), City of Hope, Duarte, CA.
  • Luo Y; Department of Diabetes Complications and Metabolism (X.T., Y.M., Y.L., K.S., F.L., C.H.L., R.N., Z.C.), City of Hope, Duarte, CA.
  • Sriram K; Department of Diabetes Complications and Metabolism (X.T., Y.M., Y.L., K.S., F.L., C.H.L., R.N., Z.C.), City of Hope, Duarte, CA.
  • Qi Z; Irell and Manella Graduate School of Biological Sciences (K.S., P.T.F., R.N., Z.C.), City of Hope, Duarte, CA.
  • Lin FM; Department of Bioengineering (Z.Q., S.Z.), University of California at San Diego, La Jolla.
  • Gu Y; Department of Diabetes Complications and Metabolism (X.T., Y.M., Y.L., K.S., F.L., C.H.L., R.N., Z.C.), City of Hope, Duarte, CA.
  • Lai CH; Department of Medicine (Y.G., K.L.P.), University of California at San Diego, La Jolla.
  • Hsu CY; Department of Diabetes Complications and Metabolism (X.T., Y.M., Y.L., K.S., F.L., C.H.L., R.N., Z.C.), City of Hope, Duarte, CA.
  • Peterson KL; Department of Internal Medicine, Taipei Medical University Hospital, Taiwan (C.Y.H).
  • Van Keuren-Jensen K; Department of Medicine (Y.G., K.L.P.), University of California at San Diego, La Jolla.
  • Fueger PT; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ (K.V.K-J.).
  • Yeo GW; Irell and Manella Graduate School of Biological Sciences (K.S., P.T.F., R.N., Z.C.), City of Hope, Duarte, CA.
  • Natarajan R; Department of Molecular and Cellular Endocrinology (P.T.F.), City of Hope, Duarte, CA.
  • Zhong S; Department of Cellular and Molecular Medicine (G.W.Y.), University of California at San Diego, La Jolla.
  • Chen ZB; Department of Diabetes Complications and Metabolism (X.T., Y.M., Y.L., K.S., F.L., C.H.L., R.N., Z.C.), City of Hope, Duarte, CA.
Circulation ; 142(4): 365-379, 2020 07 28.
Article de En | MEDLINE | ID: mdl-32393053
ABSTRACT

BACKGROUND:

Metabolic disorders such as obesity and diabetes mellitus can cause dysfunction of endothelial cells (ECs) and vascular rarefaction in adipose tissues. However, the modulatory role of ECs in adipose tissue function is not fully understood. Other than vascular endothelial growth factor-vascular endothelial growth factor receptor-mediated angiogenic signaling, little is known about the EC-derived signals in adipose tissue regulation. We previously identified Argonaute 1 (AGO1; a key component of microRNA-induced silencing complex) as a crucial regulator in hypoxia-induced angiogenesis. In this study, we intend to determine the AGO1-mediated EC transcriptome, the functional importance of AGO1-regulated endothelial function in vivo, and the relevance to adipose tissue function and obesity.

METHODS:

We generated and subjected mice with EC-AGO1 deletion (EC-AGO1-knockout [KO]) and their wild-type littermates to a fast food-mimicking, high-fat high-sucrose diet and profiled the metabolic phenotypes. We used crosslinking immunoprecipitation- and RNA-sequencing to identify the AGO1-mediated mechanisms underlying the observed metabolic phenotype of EC-AGO1-KO. We further leveraged cell cultures and mouse models to validate the functional importance of the identified molecular pathway, for which the translational relevance was explored using human endothelium isolated from healthy donors and donors with obesity/type 2 diabetes mellitus.

RESULTS:

We identified an antiobesity phenotype of EC-AGO1-KO, evident by lower body weight and body fat, improved insulin sensitivity, and enhanced energy expenditure. At the organ level, we observed the most significant phenotype in the subcutaneous and brown adipose tissues of KO mice, with greater vascularity and enhanced browning and thermogenesis. Mechanistically, EC-AGO1 suppression results in inhibition of thrombospondin-1 (THBS1/TSP1), an antiangiogenic and proinflammatory cytokine that promotes insulin resistance. In EC-AGO1-KO mice, overexpression of TSP1 substantially attenuated the beneficial phenotype. In human endothelium isolated from donors with obesity or type 2 diabetes mellitus, AGO1 and THBS1 are expressed at higher levels than the healthy controls, supporting a pathological role of this pathway.

CONCLUSIONS:

Our study suggests a novel mechanism by which ECs, through the AGO1-TSP1 pathway, control vascularization and function of adipose tissues, insulin sensitivity, and whole-body metabolic state.
Sujet(s)
Mots clés

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Tissu adipeux brun / Facteurs d'initiation eucaryotes / Prédisposition aux maladies / Endothélium / Protéines Argonaute / Maladies métaboliques Type d'étude: Diagnostic_studies / Prognostic_studies Limites: Adult / Animals / Female / Humans / Male / Middle aged Langue: En Journal: Circulation Année: 2020 Type de document: Article Pays d'affiliation: Canada

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Tissu adipeux brun / Facteurs d'initiation eucaryotes / Prédisposition aux maladies / Endothélium / Protéines Argonaute / Maladies métaboliques Type d'étude: Diagnostic_studies / Prognostic_studies Limites: Adult / Animals / Female / Humans / Male / Middle aged Langue: En Journal: Circulation Année: 2020 Type de document: Article Pays d'affiliation: Canada