3
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      CPT1A in AgRP neurons is required for sex-dependent regulation of feeding and thirst

      research-article
      1 , 2 , 1 , 2 , 3 , 4 , 1 , 2 , 1 , 2 , 3 , 5 , 6 , 6 , 7 , 1 , 2 , 1 , 2 , 8 , 9 , 10 , 1 , 11 , 1 , 11 , 12 , 1 , 11 , 12 , 3 , 13 , 3 , 13 , 8 , 9 , 10 , 3 , 14 , 3 , 4 , 15 , 16 , 17 , 16 , 17 , 1 , 2 , 3 , 1 , 2 , 3 ,
      Biology of Sex Differences
      BioMed Central
      CPT1A, Fatty acid metabolism, AgRP neurons, Energy balance, Food intake, Thirst

      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

          Background

          Fatty acid metabolism in the hypothalamus has an important role in food intake, but its specific role in AgRP neurons is poorly understood. Here, we examined whether carnitinea palmitoyltransferase 1A (CPT1A), a key enzyme in mitochondrial fatty acid oxidation, affects energy balance.

          Methods

          To obtain Cpt1aKO mice and their control littermates, Cpt1a (flox/flox) mice were crossed with tamoxifen-inducible AgRP CreERT2 mice. Food intake and body weight were analyzed weekly in both males and females. At 12 weeks of age, metabolic flexibility was determined by ghrelin-induced food intake and fasting–refeeding satiety tests. Energy expenditure was analyzed by calorimetric system and thermogenic activity of brown adipose tissue. To study fluid balance the analysis of urine and water intake volumes; osmolality of urine and plasma; as well as serum levels of angiotensin and components of RAAS (renin–angiotensin–aldosterone system) were measured. At the central level, changes in AgRP neurons were determined by: (1) analyzing specific AgRP gene expression in RiboTag– Cpt1aKO mice obtained by crossing Cpt1aKO mice with RiboTag mice; (2) measuring presynaptic terminal formation in the AgRP neurons with the injection of the AAV1 -EF1a-DIO-synaptophysin-GFP in the arcuate nucleus of the hypothalamus; (3) analyzing AgRP neuronal viability and spine formations by the injection AAV9 -EF1a-DIO-mCherry in the arcuate nucleus of the hypothalamus; (4) analyzing in situ the specific AgRP mitochondria in the ZsGreen- Cpt1aKO obtained by breeding ZsGreen mice with Cpt1aKO mice. Two-way ANOVA analyses were performed to determine the contributions of the effect of lack of CPT1A in AgRP neurons in the sex.

          Results

          Changes in food intake were just seen in male Cpt1aKO mice while only female Cpt1aKO mice increased energy expenditure. The lack of Cpt1a in the AgRP neurons enhanced brown adipose tissue activity, mainly in females, and induced a substantial reduction in fat deposits and body weight. Strikingly, both male and female Cpt1aKO mice showed polydipsia and polyuria, with more reduced serum vasopressin levels in females and without osmolality alterations, indicating a direct involvement of Cpt1a in AgRP neurons in fluid balance. AgRP neurons from Cpt1aKO mice showed a sex-dependent gene expression pattern, reduced mitochondria and decreased presynaptic innervation to the paraventricular nucleus, without neuronal viability alterations.

          Conclusions

          Our results highlight that fatty acid metabolism and CPT1A in AgRP neurons show marked sex differences and play a relevant role in the neuronal processes necessary for the maintenance of whole-body fluid and energy balance.

          Supplementary Information

          The online version contains supplementary material available at 10.1186/s13293-023-00498-8.

          Highlights

          • Fatty acid metabolism and CPT1A in AgRP neurons show marked sex-dependent differences in the control of feeding.

          • Cpt1a gene deletion in AgRP neurons increases energy expenditure in females but not in males.

          • CPT1A in AgRP neurons is involved in the control of thirst and fluid homeostasis.

          • Cpt1a gene deletion in AgRP neurons induces morphological, mitochondrial, and gene expression alterations in a sex-dependent manner.

          Supplementary Information

          The online version contains supplementary material available at 10.1186/s13293-023-00498-8.

          Related collections

          Most cited references92

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

          Cell-type-specific isolation of ribosome-associated mRNA from complex tissues.

          Gene profiling techniques allow the assay of transcripts from organs, tissues, and cells with an unprecedented level of coverage. However, most of these approaches are still limited by the fact that organs and tissues are composed of multiple cell types that are each unique in their patterns of gene expression. To identify the transcriptome from a single cell type in a complex tissue, investigators have relied upon physical methods to separate cell types or in situ hybridization and immunohistochemistry. Here, we describe a strategy to rapidly and efficiently isolate ribosome-associated mRNA transcripts from any cell type in vivo. We have created a mouse line, called RiboTag, which carries an Rpl22 allele with a floxed wild-type C-terminal exon followed by an identical C-terminal exon that has three copies of the hemagglutinin (HA) epitope inserted before the stop codon. When the RiboTag mouse is crossed to a cell-type-specific Cre recombinase-expressing mouse, Cre recombinase activates the expression of epitope-tagged ribosomal protein RPL22(HA), which is incorporated into actively translating polyribosomes. Immunoprecipitation of polysomes with a monoclonal antibody against HA yields ribosome-associated mRNA transcripts from specific cell types. We demonstrate the application of this technique in brain using neuron-specific Cre recombinase-expressing mice and in testis using a Sertoli cell Cre recombinase-expressing mouse.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Rapid, reversible activation of AgRP neurons drives feeding behavior in mice.

            Several different neuronal populations are involved in regulating energy homeostasis. Among these, agouti-related protein (AgRP) neurons are thought to promote feeding and weight gain; however, the evidence supporting this view is incomplete. Using designer receptors exclusively activated by designer drugs (DREADD) technology to provide specific and reversible regulation of neuronal activity in mice, we have demonstrated that acute activation of AgRP neurons rapidly and dramatically induces feeding, reduces energy expenditure, and ultimately increases fat stores. All these effects returned to baseline after stimulation was withdrawn. In contrast, inhibiting AgRP neuronal activity in hungry mice reduced food intake. Together, these findings demonstrate that AgRP neuron activity is both necessary and sufficient for feeding. Of interest, activating AgRP neurons potently increased motivation for feeding and also drove intense food-seeking behavior, demonstrating that AgRP neurons engage brain sites controlling multiple levels of feeding behavior. Due to its ease of use and suitability for both acute and chronic regulation, DREADD technology is ideally suited for investigating the neural circuits hypothesized to regulate energy balance.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Insulin action in AgRP-expressing neurons is required for suppression of hepatic glucose production.

              Insulin action in the central nervous system regulates energy homeostasis and glucose metabolism. To define the insulin-responsive neurons that mediate these effects, we generated mice with selective inactivation of the insulin receptor (IR) in either pro-opiomelanocortin (POMC)- or agouti-related peptide (AgRP)-expressing neurons of the arcuate nucleus of the hypothalamus. While neither POMC- nor AgRP-restricted IR knockout mice exhibited altered energy homeostasis, insulin failed to normally suppress hepatic glucose production during euglycemic-hyperinsulinemic clamps in AgRP-IR knockout (IR(DeltaAgRP)) mice. These mice also exhibited reduced insulin-stimulated hepatic interleukin-6 expression and increased hepatic expression of glucose-6-phosphatase. These results directly demonstrate that insulin action in POMC and AgRP cells is not required for steady-state regulation of food intake and body weight. However, insulin action specifically in AgRP-expressing neurons does play a critical role in controlling hepatic glucose production and may provide a target for the treatment of insulin resistance in type 2 diabetes.
                Bookmark

                Author and article information

                Contributors
                dserra@ub.edu
                Journal
                Biol Sex Differ
                Biol Sex Differ
                Biology of Sex Differences
                BioMed Central (London )
                2042-6410
                25 March 2023
                25 March 2023
                2023
                : 14
                : 14
                Affiliations
                [1 ]GRID grid.5841.8, ISNI 0000 0004 1937 0247, Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, , Universitat de Barcelona, ; Av. Joan XXIII, 27-31, 08028 Barcelona, Spain
                [2 ]GRID grid.5841.8, ISNI 0000 0004 1937 0247, Institut de Biomedicina de la Universitat de Barcelona (IBUB), , Universitat de Barcelona, ; Barcelona, Spain
                [3 ]GRID grid.413448.e, ISNI 0000 0000 9314 1427, Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), , Instituto de Salud Carlos III, ; Madrid, Spain
                [4 ]GRID grid.11794.3a, ISNI 0000000109410645, NeurObesity Group, Department of Physiology, , CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, ; Santiago de Compostela, Spain
                [5 ]GRID grid.10403.36, ISNI 0000000091771775, Neuronal Control of Metabolism Laboratory, , Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), ; Barcelona, Spain
                [6 ]GRID grid.11478.3b, ISNI 0000 0004 1766 3695, CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, ; 08028 Barcelona, Spain
                [7 ]GRID grid.5612.0, ISNI 0000 0001 2172 2676, Universitat Pompeu Fabra (UPF), ; Barcelona, Spain
                [8 ]GRID grid.420258.9, ISNI 0000 0004 1794 1077, Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), ; Barcelona, Spain
                [9 ]GRID grid.413448.e, ISNI 0000 0000 9314 1427, CIBER de Enfermedades Cardiovasculares (CIBERCV), , Instituto de Salud Carlos III, ; Madrid, Spain
                [10 ]Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), Barcelona, Spain
                [11 ]GRID grid.418264.d, ISNI 0000 0004 1762 4012, Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), ; Madrid, Spain
                [12 ]GRID grid.5841.8, ISNI 0000 0004 1937 0247, Institute of Neurosciences of the Universitat de Barcelona, ; Barcelona, Spain
                [13 ]GRID grid.5841.8, ISNI 0000 0004 1937 0247, Department of Inorganic & Organic Chemistry, Faculty of Chemistry, , Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, ; Barcelona, Spain
                [14 ]GRID grid.410675.1, ISNI 0000 0001 2325 3084, Department of Basic Sciences, Faculty of Medicine & Health Sciences, , Universitat Internacional de Catalunya, ; Sant Cugat del Vallès, Spain
                [15 ]GRID grid.34477.33, ISNI 0000000122986657, Department of Biochemistry, , Howard Hughes Medical Institute, University of Washington, ; Seattle, WA USA
                [16 ]GRID grid.7080.f, ISNI 0000 0001 2296 0625, Department of Cell Biology, Physiology and Immunology, , Universitat Autònoma de Barcelona, ; Bellaterra, Spain
                [17 ]GRID grid.7080.f, ISNI 0000 0001 2296 0625, Institut de Neurociències, , Universitat Autònoma de Barcelona, ; Bellaterra, Spain
                Author information
                http://orcid.org/0000-0002-4936-4206
                Article
                498
                10.1186/s13293-023-00498-8
                10040140
                36966335
                3b4500da-f894-4aef-b445-c26cd74d8f5c
                © The Author(s) 2023

                Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

                History
                : 26 October 2022
                : 10 March 2023
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/501100003329, Ministerio de Economía y Competitividad;
                Award ID: SAF2017-83813-C3-1-R
                Award ID: SAF2017-88108-R
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100004837, Ministerio de Ciencia e Innovación;
                Award ID: RTI2018-094727-B-100
                Award ID: PID2020-114953RB-C21
                Award ID: PID2020-114977RB-I00
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100019180, HORIZON EUROPE European Research Council;
                Award ID: ERC-2014-StG-638106
                Award Recipient :
                Categories
                Research
                Custom metadata
                © The Author(s) 2023

                Human biology
                cpt1a,fatty acid metabolism,agrp neurons,energy balance,food intake,thirst
                Human biology
                cpt1a, fatty acid metabolism, agrp neurons, energy balance, food intake, thirst

                Comments

                Comment on this article