41
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Reconstitution of UCP1 using CRISPR/Cas9 in the white adipose tissue of pigs decreases fat deposition and improves thermogenic capacity

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      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.

          Significance

          Uncoupling protein 1 (UCP1) is responsible for brown adipose tissue-mediated thermogenesis and plays a critical role in protecting against cold and regulating energy homeostasis. Modern pigs lack functional UCP1, which makes them susceptible to cold and prone to fat deposition and results in neonatal mortality and decreased production efficiency. In the current study, a CRISPR/Cas9-mediated homologous recombination-independent approach was established, and mouse adiponectin-UCP1 was efficiently inserted into the porcine endogenous UCP1 locus. The resultant UCP1 KI pigs showed an improved ability to maintain body temperature, decreased fat deposition, and increased carcass lean percentage. UCP1 KI pigs are a potentially valuable resource for the pig industry that can improve pig welfare and reduce economic losses.

          Abstract

          Uncoupling protein 1 (UCP1) is localized on the inner mitochondrial membrane and generates heat by uncoupling ATP synthesis from proton transit across the inner membrane. UCP1 is a key element of nonshivering thermogenesis and is most likely important in the regulation of body adiposity. Pigs (Artiodactyl family Suidae) lack a functional UCP1 gene, resulting in poor thermoregulation and susceptibility to cold, which is an economic and pig welfare issue owing to neonatal mortality. Pigs also have a tendency toward fat accumulation, which may be linked to their lack of UCP1, and thus influences the efficiency of pig production. Here, we report application of a CRISPR/Cas9-mediated, homologous recombination (HR)-independent approach to efficiently insert mouse adiponectin-UCP1 into the porcine endogenous UCP1 locus. The resultant UCP1 knock-in (KI) pigs showed an improved ability to maintain body temperature during acute cold exposure, but they did not have alterations in physical activity levels or total daily energy expenditure (DEE). Furthermore, ectopic UCP1 expression in white adipose tissue (WAT) dramatically decreased fat deposition by 4.89% ( P < 0.01), consequently increasing carcass lean percentage (CLP; P < 0.05). Mechanism studies indicated that the loss of fat upon UCP1 activation in WAT was linked to elevated lipolysis. UCP1 KI pigs are a potentially valuable resource for agricultural production through their combination of cold adaptation, which improves pig welfare and reduces economic losses, with reduced fat deposition and increased lean meat production.

          Related collections

          Most cited references46

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

          Mice lacking mitochondrial uncoupling protein are cold-sensitive but not obese.

          The mitochondrial uncoupling protein (UCP) in the mitochondrial inner membrane of mammalian brown adipose tissue generates heat by uncoupling oxidative phosphorylation. This process protects against cold and regulates energy balance. Manipulation of thermogenesis could be an effective strategy against obesity. Here we determine the role of UCP in the regulation of body mass by targeted inactivation of the gene encoding it. We find that UCP-deficient mice consume less oxygen after treatment with a beta3-adrenergic-receptor agonist and that they are sensitive to cold, indicating that their thermoregulation is defective. However, this deficiency caused neither hyperphagia nor obesity in mice fed on either a standard or a high-fat diet. We propose that the loss of UCP may be compensated by UCP2, a newly discovered homologue of UCP; this gene is ubiquitously expressed and is induced in the brown fat of UCP-deficient mice.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Brown remodeling of white adipose tissue by SirT1-dependent deacetylation of Pparγ.

            Brown adipose tissue (BAT) can disperse stored energy as heat. Promoting BAT-like features in white adipose (WAT) is an attractive, if elusive, therapeutic approach to staunch the current obesity epidemic. Here we report that gain of function of the NAD-dependent deacetylase SirT1 or loss of function of its endogenous inhibitor Deleted in breast cancer-1 (Dbc1) promote "browning" of WAT by deacetylating peroxisome proliferator-activated receptor (Ppar)-γ on Lys268 and Lys293. SirT1-dependent deacetylation of Lys268 and Lys293 is required to recruit the BAT program coactivator Prdm16 to Pparγ, leading to selective induction of BAT genes and repression of visceral WAT genes associated with insulin resistance. An acetylation-defective Pparγ mutant induces a brown phenotype in white adipocytes, whereas an acetylated mimetic fails to induce "brown" genes but retains the ability to activate "white" genes. We propose that SirT1-dependent Pparγ deacetylation is a form of selective Pparγ modulation of potential therapeutic import. Copyright © 2012 Elsevier Inc. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Brown adipose tissue as a secretory organ.

              Brown adipose tissue (BAT) is the main site of adaptive thermogenesis and experimental studies have associated BAT activity with protection against obesity and metabolic diseases, such as type 2 diabetes mellitus and dyslipidaemia. Active BAT is present in adult humans and its activity is impaired in patients with obesity. The ability of BAT to protect against chronic metabolic disease has traditionally been attributed to its capacity to utilize glucose and lipids for thermogenesis. However, BAT might also have a secretory role, which could contribute to the systemic consequences of BAT activity. Several BAT-derived molecules that act in a paracrine or autocrine manner have been identified. Most of these factors promote hypertrophy and hyperplasia of BAT, vascularization, innervation and blood flow, processes that are all associated with BAT recruitment when thermogenic activity is enhanced. Additionally, BAT can release regulatory molecules that act on other tissues and organs. This secretory capacity of BAT is thought to be involved in the beneficial effects of BAT transplantation in rodents. Fibroblast growth factor 21, IL-6 and neuregulin 4 are among the first BAT-derived endocrine factors to be identified. In this Review, we discuss the current understanding of the regulatory molecules (the so-called brown adipokines or batokines) that are released by BAT that influence systemic metabolism and convey the beneficial metabolic effects of BAT activation. The identification of such adipokines might also direct drug discovery approaches for managing obesity and its associated chronic metabolic diseases.
                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc. Natl. Acad. Sci. U.S.A
                pnas
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                7 November 2017
                23 October 2017
                : 114
                : 45
                : E9474-E9482
                Affiliations
                [1] aState Key Laboratory of Stem Cell and Reproductive Biology, Chinese Academy of Sciences, Chaoyang District, Beijing, China 100101;
                [2] bSavaid Medical School, University of Chinese Academy of Sciences, Beijing, China 100049;
                [3] cKey Laboratory of Animal Ecology and Conservation Biology, Chinese Academy of Sciences, Chaoyang District, Beijing, China 100101;
                [4] dCollege of Life Science, University of Chinese Academy of Sciences, Beijing, China 100049;
                [5] eInstitute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China 100101;
                [6] fEnergetics Research Group, Zoology Department, University of Aberdeen, Aberdeen AB24 2TZ, United Kingdom;
                [7] gDepartment of Animal Science, Yanbian University, Yanji, Jilin, China 133002;
                [8] hInstitute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China 100193
                Author notes
                2To whom correspondence may be addressed. Email: wangyanfang@ 123456caas.cn , jinw@ 123456ioz.ac.cn , or zhaojg@ 123456ioz.ac.cn .

                Edited by R. Michael Roberts, University of Missouri-Columbia, Columbia, MO, and approved September 14, 2017 (received for review May 12, 2017)

                Author contributions: Q. Zhou, Y.W., W.J., and J.Z. designed research; Q. Zheng, J.L., J.H., H.Z., R.Z., X.Z., C.H., G.Q., J.Y., R.S., Q.J., X.W., Y.L., N.Z., Z.P., R.Y., J.R.S., and H.W. performed research; Q. Zheng, J.L., X.Z., C.C., J.R.S., H.W., Q. Zhou, Y.W., and J.Z. analyzed data; and Q. Zheng, J.R.S., Y.W., and J.Z. wrote the paper.

                1Q. Zheng, J.L., and J.H. contributed equally to this work.

                Author information
                http://orcid.org/0000-0001-6587-4823
                Article
                PMC5692550 PMC5692550 5692550 201707853
                10.1073/pnas.1707853114
                5692550
                29078316
                23f51852-7704-41fa-98d5-d161f2af064c
                Published under the PNAS license.
                History
                Page count
                Pages: 9
                Funding
                Funded by: National transgenic project of China
                Award ID: 2016ZX08009003-006-007
                Funded by: The Strategic Priority Research Programs of CAS
                Award ID: XDA08010304
                Funded by: The Strategic Priority Research of CAS
                Award ID: XDB13030000
                Funded by: The National Natural Science Foundation of China
                Award ID: 81671274
                Funded by: The National Natural Science Foundation of China
                Award ID: 31272440
                Funded by: The National Program on Key Basic Research Project
                Award ID: 2015CB943100
                Categories
                PNAS Plus
                Biological Sciences
                Agricultural Sciences
                PNAS Plus

                fat deposition,UCP1,thermoregulation,pig,CRISPR/Cas9
                fat deposition, UCP1, thermoregulation, pig, CRISPR/Cas9

                Comments

                Comment on this article