17
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Activation of NF-κB drives the enhanced survival of adipose tissue macrophages in an obesogenic environment

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Objective

          Macrophage accumulation in adipose tissue (AT) during obesity contributes to inflammation and insulin resistance. Recruitment of monocytes to obese AT has been the most studied mechanism explaining this accumulation. However, recent evidence suggests that recruitment-independent mechanisms may also regulate pro-inflammatory AT macrophage (ATM) numbers. The role of increased ATM survival during obesity has yet to be explored.

          Results

          We demonstrate that activation of apoptotic pathways is significantly reduced in ATMs from diet-induced and genetically obese mice. Concurrently, pro-survival Bcl-2 family member protein levels and localization to the mitochondria is elevated in ATMs from obese mice. This increased pro-survival signaling was associated with elevated activation of the transcription factor, NF-κB, and increased expression of its pro-survival target genes. Finally, an obesogenic milieu increased ATM viability only when NF-κB signaling pathways were functional.

          Conclusions

          Our data demonstrate that obesity promotes survival of inflammatory ATMs, possibly through an NF-κB-regulated mechanism.

          Graphical abstract

          Highlights

          • Macrophage apoptosis is decreased in obese adipose tissue.

          • ATMs from obese mice display increased mitochondrial localization of Bcl-2.

          • The pro-survival targets of NF-κB are increased in ATMs from obese mice.

          • NF-κB activation in ATMs during metabolic stimulation increases their survival.

          • Decreased ATM apoptosis contributes to macrophage accumulation in obesity.

          Related collections

          Most cited references46

          • Record: found
          • Abstract: found
          • Article: not found

          MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity.

          Adipocytes secrete a variety of bioactive molecules that affect the insulin sensitivity of other tissues. We now show that the abundance of monocyte chemoattractant protein-1 (MCP-1) mRNA in adipose tissue and the plasma concentration of MCP-1 were increased both in genetically obese diabetic (db/db) mice and in WT mice with obesity induced by a high-fat diet. Mice engineered to express an MCP-1 transgene in adipose tissue under the control of the aP2 gene promoter exhibited insulin resistance, macrophage infiltration into adipose tissue, and increased hepatic triglyceride content. Furthermore, insulin resistance, hepatic steatosis, and macrophage accumulation in adipose tissue induced by a high-fat diet were reduced extensively in MCP-1 homozygous KO mice compared with WT animals. Finally, acute expression of a dominant-negative mutant of MCP-1 ameliorated insulin resistance in db/db mice and in WT mice fed a high-fat diet. These findings suggest that an increase in MCP-1 expression in adipose tissue contributes to the macrophage infiltration into this tissue, insulin resistance, and hepatic steatosis associated with obesity in mice.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Macrophages, inflammation, and insulin resistance.

            Obesity induces an insulin-resistant state in adipose tissue, liver, and muscle and is a strong risk factor for the development of type 2 diabetes mellitus. Insulin resistance in the setting of obesity results from a combination of altered functions of insulin target cells and the accumulation of macrophages that secrete proinflammatory mediators. At the molecular level, insulin resistance is promoted by a transition in macrophage polarization from an alternative M2 activation state maintained by STAT6 and PPARs to a classical M1 activation state driven by NF-kappaB, AP1, and other signal-dependent transcription factors that play crucial roles in innate immunity. Strategies focused on inhibiting the inflammation/insulin resistance axis that otherwise preserve essential innate immune functions may hold promise for therapeutic intervention.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Metabolic dysfunction drives a mechanistically distinct proinflammatory phenotype in adipose tissue macrophages.

              Adipose tissue macrophage (ATM)-driven inflammation plays a key role in insulin resistance; however, factors activating ATMs are poorly understood. Using a proteomics approach, we show that markers of classical activation are absent on ATMs from obese humans but are readily detectable on airway macrophages of patients with cystic fibrosis, a disease associated with chronic bacterial infection. Moreover, treating macrophages with glucose, insulin, and palmitate-conditions characteristic of the metabolic syndrome-produces a "metabolically activated" phenotype distinct from classical activation. Markers of metabolic activation are expressed by proinflammatory ATMs in obese humans/mice and are positively correlated with adiposity. Metabolic activation is driven by independent proinflammatory and anti-inflammatory pathways, which regulate balance between cytokine production and lipid metabolism. We identify PPARγ and p62/SQSTM1 as two key proteins that promote lipid metabolism and limit inflammation in metabolically activated macrophages. Collectively, our data provide important mechanistic insights into pathways that drive the metabolic-disease-specific phenotype of macrophages.
                Bookmark

                Author and article information

                Contributors
                Journal
                Mol Metab
                Mol Metab
                Molecular Metabolism
                Elsevier
                2212-8778
                28 July 2015
                October 2015
                28 July 2015
                : 4
                : 10
                : 665-677
                Affiliations
                [1 ]Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, United States
                [2 ]Department of Veterans Affairs, Tennessee Valley Healthcare System, United States
                [3 ]Department of Cancer Biology, Vanderbilt University School of Medicine, United States
                Author notes
                []Corresponding author. Vanderbilt University School of Medicine, Room 702 Light Hall, Nashville, TN 37232-0615, United States. Tel.: +1 615 322 5177. alyssa.hasty@ 123456vanderbilt.edu
                [4]

                Department of Medicine, Indiana University School of Medicine, United States.

                [5]

                Andrea A. Hill and Emily K. Anderson-Baucum contributed equally to this work.

                Article
                S2212-8778(15)00139-8
                10.1016/j.molmet.2015.07.005
                4588436
                26779432
                671475fa-decf-4516-8f57-420a1e53ff09

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 23 June 2015
                : 10 July 2015
                : 18 July 2015
                Categories
                Original Article

                adipose tissue,macrophage,apoptosis,cleaved caspase-3,nf-κb,survival

                Comments

                Comment on this article