File Download
  Links for fulltext
     (May Require Subscription)
Supplementary

postgraduate thesis: Adipose miR-34a in obesity-induced metabolic inflammation and insulin resistance

TitleAdipose miR-34a in obesity-induced metabolic inflammation and insulin resistance
Authors
Issue Date2018
PublisherThe University of Hong Kong (Pokfulam, Hong Kong)
Citation
Pan, Y. [潘勇]. (2018). Adipose miR-34a in obesity-induced metabolic inflammation and insulin resistance. (Thesis). University of Hong Kong, Pokfulam, Hong Kong SAR.
AbstractAdipose tissue macrophages (ATMs) are central to local and systemic inflammation. Upon obesity, ATMs are more prone to polarize to the pro-inflammatory M1 subtype, whereas the anti-inflammatory M2 profile is suppressed. These two events collectively lead to adipose tissue inflammation. However, the underlying mechanisms explaining macrophage polarization are not fully explored. Furthermore, a clinical study has detected altered expression of microRNA (miR)-34a in obese subcutaneous adipose tissue, but whether adipose-originated miR-34a modulates obesity-associated adipose tissue remodeling and whole body metabolism remains elusive. Therefore, this study aims to investigate: 1) whether adipose miR-34a mediates adipose tissue inflammatory response and systemic insulin insensitivity via affecting macrophage polarization; 2) the potential mechanisms linking adipose miR-34a and macrophage polarization; 3) how adipocytes and macrophages crosstalk with each other under obese condition. Our results show that high fat diet (HFD) and inflammatory cues cause a strong elevation of miR-34a in adipose tissues. Adipose tissue-specific depletion of miR-34a protects mice from high fat diet-induced glucose intolerance, insulin insensitivity and other metabolic abnormalities. Macrophage depletion experiment demonstrated that the metabolic benefits in adipose-restricted miR-34a knockout mice are mainly attributed to its modulation on adipose macrophages. Indeed, miR-34a deficiency in adipose tissue leads to reduced number of crown-like structures, and increased macrophage polarization toward the anti-inflammatory M2 subtypes. Luciferase assay determined that miR-34a directly targets 3’-UTR of Kruppel-like factor 4 (Klf4). Lentivirus-mediated knocking-down of Klf4 in miR-34a knockout mice leads to exacerbated macrophage infiltration, pro-inflammatory phenotypes of macrophages in vivo, while overexpression of Klf4 improves diet-induced adipose inflammation. More importantly, adipocyte miR-34a modulates macrophage polarization in a paracrine manner via transportation of exosome. Furthermore, a strong and positive association is revealed between visceral adipose tissue miR-34a and inflammation in overweight/obese human individuals. Generally, our findings demonstrate adipose miR-34a polarizes macrophages to M1 profile and induces insulin resistance. Furthermore, the miR-34a/KLF4 axis, exhibiting as the crosstalk between adipocytes and macrophages, constitutes a critical branch in the regulation of ATM and systemic metabolic homeostasis.
DegreeDoctor of Philosophy
SubjectAdipose tissues
MicroRNA
Insulin resistance
Metabolic syndrome
Obesity - Complications
Dept/ProgramMedicine
Persistent Identifierhttp://hdl.handle.net/10722/267328

 

DC FieldValueLanguage
dc.contributor.authorPan, Yong-
dc.contributor.author潘勇-
dc.date.accessioned2019-02-18T08:45:42Z-
dc.date.available2019-02-18T08:45:42Z-
dc.date.issued2018-
dc.identifier.citationPan, Y. [潘勇]. (2018). Adipose miR-34a in obesity-induced metabolic inflammation and insulin resistance. (Thesis). University of Hong Kong, Pokfulam, Hong Kong SAR.-
dc.identifier.urihttp://hdl.handle.net/10722/267328-
dc.description.abstractAdipose tissue macrophages (ATMs) are central to local and systemic inflammation. Upon obesity, ATMs are more prone to polarize to the pro-inflammatory M1 subtype, whereas the anti-inflammatory M2 profile is suppressed. These two events collectively lead to adipose tissue inflammation. However, the underlying mechanisms explaining macrophage polarization are not fully explored. Furthermore, a clinical study has detected altered expression of microRNA (miR)-34a in obese subcutaneous adipose tissue, but whether adipose-originated miR-34a modulates obesity-associated adipose tissue remodeling and whole body metabolism remains elusive. Therefore, this study aims to investigate: 1) whether adipose miR-34a mediates adipose tissue inflammatory response and systemic insulin insensitivity via affecting macrophage polarization; 2) the potential mechanisms linking adipose miR-34a and macrophage polarization; 3) how adipocytes and macrophages crosstalk with each other under obese condition. Our results show that high fat diet (HFD) and inflammatory cues cause a strong elevation of miR-34a in adipose tissues. Adipose tissue-specific depletion of miR-34a protects mice from high fat diet-induced glucose intolerance, insulin insensitivity and other metabolic abnormalities. Macrophage depletion experiment demonstrated that the metabolic benefits in adipose-restricted miR-34a knockout mice are mainly attributed to its modulation on adipose macrophages. Indeed, miR-34a deficiency in adipose tissue leads to reduced number of crown-like structures, and increased macrophage polarization toward the anti-inflammatory M2 subtypes. Luciferase assay determined that miR-34a directly targets 3’-UTR of Kruppel-like factor 4 (Klf4). Lentivirus-mediated knocking-down of Klf4 in miR-34a knockout mice leads to exacerbated macrophage infiltration, pro-inflammatory phenotypes of macrophages in vivo, while overexpression of Klf4 improves diet-induced adipose inflammation. More importantly, adipocyte miR-34a modulates macrophage polarization in a paracrine manner via transportation of exosome. Furthermore, a strong and positive association is revealed between visceral adipose tissue miR-34a and inflammation in overweight/obese human individuals. Generally, our findings demonstrate adipose miR-34a polarizes macrophages to M1 profile and induces insulin resistance. Furthermore, the miR-34a/KLF4 axis, exhibiting as the crosstalk between adipocytes and macrophages, constitutes a critical branch in the regulation of ATM and systemic metabolic homeostasis. -
dc.languageeng-
dc.publisherThe University of Hong Kong (Pokfulam, Hong Kong)-
dc.relation.ispartofHKU Theses Online (HKUTO)-
dc.rightsThe author retains all proprietary rights, (such as patent rights) and the right to use in future works.-
dc.rightsThis work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.-
dc.subject.lcshAdipose tissues-
dc.subject.lcshMicroRNA-
dc.subject.lcshInsulin resistance-
dc.subject.lcshMetabolic syndrome-
dc.subject.lcshObesity - Complications-
dc.titleAdipose miR-34a in obesity-induced metabolic inflammation and insulin resistance-
dc.typePG_Thesis-
dc.description.thesisnameDoctor of Philosophy-
dc.description.thesislevelDoctoral-
dc.description.thesisdisciplineMedicine-
dc.description.naturepublished_or_final_version-
dc.identifier.doi10.5353/th_991044019483703414-
dc.date.hkucongregation2018-
dc.identifier.mmsid991044019483703414-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats