1
|
Ali A, Zhang ZD, Gao T, Aleksic S, Gavathiotis E, Barzilai N, Milman S. Identification of functional rare coding variants in IGF-1 gene in humans with exceptional longevity. Sci Rep 2025; 15:10199. [PMID: 40133344 PMCID: PMC11937401 DOI: 10.1038/s41598-025-94094-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
Diminished signaling via insulin/insulin-like growth factor-1 (IGF-1) axis is associated with longevity in different model organisms. IGF-1 gene is highly conserved across species, with only few evolutionary changes identified in it. Despite its potential role in regulating lifespan, no coding variants in IGF-1 have been reported in human longevity cohorts to date. This study investigated the whole exome sequencing data from 2,108 individuals in a cohort of Ashkenazi Jewish centenarians, their offspring, and controls without familial longevity to identify functional IGF-1 coding variants. We identified two likely functional coding variants IGF-1:p.Ile91Leu and IGF-1:p.Ala118Thr in our longevity cohort. Notably, a centenarian specific novel variant IGF-1:p.Ile91Leu was located at the binding interface of IGF-1-IGF-1R, whereas IGF-1:p.Ala118Thr was significantly associated with lower circulating levels of IGF-1. We performed extended all-atom molecular dynamics simulations to evaluate the impact of Ile91Leu on stability, binding dynamics and energetics of IGF-1 bound to IGF-1R. The IGF-1:p.Ile91Leu formed less stable interactions with IGF-1R's critical binding pocket residues and demonstrated lower binding affinity at the extracellular binding site compared to wild-type IGF-1. Our findings suggest that IGF-1:p.Ile91Leu and IGF-1:p.Ala118Thr variants attenuate IGF-1R activity by impairing IGF-1 binding and diminishing the circulatory levels of IGF-1, respectively. Consequently, diminished IGF-1 signaling resulting from these variants may contribute to exceptional longevity in humans.
Collapse
Affiliation(s)
- Amanat Ali
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA.
- Institute for Aging Research and the Einstein-NSC, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA.
| | - Zhengdong D Zhang
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA
- Institute for Aging Research and the Einstein-NSC, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA
| | - Tina Gao
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA
- Institute for Aging Research and the Einstein-NSC, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA
| | - Sandra Aleksic
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA
- Institute for Aging Research and the Einstein-NSC, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA
| | - Evripidis Gavathiotis
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA
- Institute for Aging Research and the Einstein-NSC, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA
| | - Nir Barzilai
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA
- Institute for Aging Research and the Einstein-NSC, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA
| | - Sofiya Milman
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA.
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA.
- Institute for Aging Research and the Einstein-NSC, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA.
| |
Collapse
|
2
|
Force E, Alvarez C, Fuentes A, Maria A, Bozzolan F, Debernard S. Diet influence on male sexual maturation through interplay between insulin signaling and juvenile hormone in insects. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2025; 177:104252. [PMID: 39701395 DOI: 10.1016/j.ibmb.2024.104252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/28/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
In animals, sexual maturation coincides with the development of sexual behaviors and reproductive system. These developmental events are influenced by diet and governed by endocrine signals. Here, for the first time in insects, we explored functional links between nutrition and juvenile hormone (JH) in the male reproductive physiology through the insulin signaling pathway (ISP) acting as a transducer of nutritional signals. We turned to the male moth Agrotis ipsilon for which sexual maturation, including accessory sex glands (ASGs) development concomitantly with antennal lobes (ALs) maturation for female sex pheromone processing and display of sexual behavior, is known to be JH- and diet-dependent. Indeed, a diet rich in sugars with sodium was previously shown to accelerate sexual maturation, which was achieved from the third day of adult life. In this study, we demonstrated that such a diet raised i) the expression of JH signaling actors (Methoprene-tolerant, Taiman, and Krüppel homolog 1) in ALs and ASGs, ii) the biosynthesis and circulating levels of JH, and iii) the expression of both insulin receptor (InR) and insulin-like peptides (ILPs) in corpora allata (CAs) and brain respectively. Insulin injection raised JH biosynthesis following increased HMG-CoA reductase expression in CAs; opposite effects were induced in InR-deficient males. Thus, we highlighted that promoting effects of a diet composed of sugars with sodium on male sexual maturation results from an early induction of ISP causing an increase in JH biosynthesis followed by a potentiation of JH actions on the development of ASGs and ALs in A. ipsilon.
Collapse
Affiliation(s)
- Evan Force
- Sorbonne Université, Université Paris-Est Créteil, INRAE, CNRS, IRD, Institute for Ecology and Environmental Sciences of Paris, France.
| | | | - Annabelle Fuentes
- Sorbonne Université, Université Paris-Est Créteil, INRAE, CNRS, IRD, Institute for Ecology and Environmental Sciences of Paris, France
| | - Annick Maria
- Sorbonne Université, Université Paris-Est Créteil, INRAE, CNRS, IRD, Institute for Ecology and Environmental Sciences of Paris, France
| | - Françoise Bozzolan
- Sorbonne Université, Université Paris-Est Créteil, INRAE, CNRS, IRD, Institute for Ecology and Environmental Sciences of Paris, France
| | - Stéphane Debernard
- Sorbonne Université, Université Paris-Est Créteil, INRAE, CNRS, IRD, Institute for Ecology and Environmental Sciences of Paris, France.
| |
Collapse
|
3
|
Gluth A, Li X, Gritsenko MA, Gaffrey MJ, Kim DN, Lalli PM, Chu RK, Day NJ, Sagendorf TJ, Monroe ME, Feng S, Liu T, Yang B, Qian WJ, Zhang T. Integrative Multi-PTM Proteomics Reveals Dynamic Global, Redox, Phosphorylation, and Acetylation Regulation in Cytokine-Treated Pancreatic Beta Cells. Mol Cell Proteomics 2024; 23:100881. [PMID: 39550035 PMCID: PMC11700301 DOI: 10.1016/j.mcpro.2024.100881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/28/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024] Open
Abstract
Studying regulation of protein function at a systems level necessitates an understanding of the interplay among diverse posttranslational modifications (PTMs). A variety of proteomics sample processing workflows are currently used to study specific PTMs but rarely characterize multiple types of PTMs from the same sample inputs. Method incompatibilities and laborious sample preparation steps complicate large-scale physiological investigations and can lead to variations in results. The single-pot, solid-phase-enhanced sample preparation (SP3) method for sample cleanup is compatible with different lysis buffers and amenable to automation, making it attractive for high-throughput multi-PTM profiling. Herein, we describe an integrative SP3 workflow for multiplexed quantification of protein abundance, cysteine thiol oxidation, phosphorylation, and acetylation. The broad applicability of this approach is demonstrated using cell and tissue samples, and its utility for studying interacting regulatory networks is highlighted in a time-course experiment of cytokine-treated β-cells. We observed a swift response in the global regulation of protein abundances consistent with rapid activation of JAK-STAT and NF-κB signaling pathways. Regulators of these pathways as well as proteins involved in their target processes displayed multi-PTM dynamics indicative of complex cellular response stages: acute, adaptation, and chronic (prolonged stress). PARP14, a negative regulator of JAK-STAT, had multiple colocalized PTMs that may be involved in intraprotein regulatory crosstalk. Our workflow provides a high-throughput platform that can profile multi-PTMomes from the same sample set, which is valuable in unraveling the functional roles of PTMs and their co-regulation.
Collapse
Affiliation(s)
- Austin Gluth
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA; Department of Biological Systems Engineering, Washington State University, Richland, Washington, USA
| | - Xiaolu Li
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Marina A Gritsenko
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Matthew J Gaffrey
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Doo Nam Kim
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Priscila M Lalli
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Rosalie K Chu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Nicholas J Day
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Tyler J Sagendorf
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Matthew E Monroe
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Song Feng
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Tao Liu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Bin Yang
- Department of Biological Systems Engineering, Washington State University, Richland, Washington, USA
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Tong Zhang
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA.
| |
Collapse
|
4
|
Jiang BC, Ling YJ, Xu ML, Gu J, Wu XB, Sha WL, Tian T, Bai XH, Li N, Jiang CY, Chen O, Ma LJ, Zhang ZJ, Qin YB, Zhu M, Yuan HJ, Wu LJ, Ji RR, Gao YJ. Follistatin drives neuropathic pain in mice through IGF1R signaling in nociceptive neurons. Sci Transl Med 2024; 16:eadi1564. [PMID: 39413164 DOI: 10.1126/scitranslmed.adi1564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/10/2024] [Accepted: 09/25/2024] [Indexed: 10/18/2024]
Abstract
Neuropathic pain is a debilitating chronic condition that lacks effective treatment. The role of cytokine- and chemokine-mediated neuroinflammation in its pathogenesis has been well documented. Follistatin (FST) is a secreted protein known to antagonize the biological activity of cytokines in the transforming growth factor-β (TGF-β) superfamily. The involvement of FST in neuropathic pain and the underlying mechanism remain largely unknown. Here, we report that FST was up-regulated in A-fiber sensory neurons after spinal nerve ligation (SNL) in mice. Inhibition or deletion of FST alleviated neuropathic pain and reduced the nociceptive neuron hyperexcitability induced by SNL. Conversely, intrathecal or intraplantar injection of recombinant FST, or overexpression of FST in the dorsal root ganglion (DRG) neurons, induced pain hypersensitivity. Furthermore, exogenous FST increased neuronal excitability in nociceptive neurons. The biolayer interferometry (BLI) assay and coimmunoprecipitation (co-IP) demonstrated direct binding of FST to the insulin-like growth factor-1 receptor (IGF1R), and IGF1R inhibition reduced FST-induced activation of extracellular signal-regulated kinase (ERK) and protein kinase B (AKT), as well as neuronal hyperexcitability. Further co-IP analysis revealed that the N-terminal domain of FST exhibits the highest affinity for IGF1R, and blocking this interaction with a peptide derived from FST attenuated Nav1.7-mediated neuronal hyperexcitability and neuropathic pain after SNL. In addition, FST enhanced neuronal excitability in human DRG neurons through IGF1R. Collectively, our findings suggest that FST, released from A-fiber neurons, enhances Nav1.7-mediated hyperexcitability of nociceptive neurons by binding to IGF1R, making it a potential target for neuropathic pain treatment.
Collapse
Affiliation(s)
- Bao-Chun Jiang
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Yue-Juan Ling
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Meng-Lin Xu
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Jun Gu
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Xiao-Bo Wu
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Wei-Lin Sha
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Tian Tian
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Xue-Hui Bai
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Nan Li
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong 518052, China
| | - Chang-Yu Jiang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong 518052, China
| | - Ouyang Chen
- Center for Translational Pain Medicine, Departments of Anesthesiology, Cell Biology, and Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ling-Jie Ma
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Zhi-Jun Zhang
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Yi-Bin Qin
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Meixuan Zhu
- Department of Psychology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hong-Jie Yuan
- Department of Pain Management, Nantong Hospital of Traditional Chinese Medicine, Jiangsu 226001, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Departments of Anesthesiology, Cell Biology, and Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yong-Jing Gao
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| |
Collapse
|
5
|
Millward DJ. Post-natal muscle growth and protein turnover: a narrative review of current understanding. Nutr Res Rev 2024; 37:141-168. [PMID: 37395180 DOI: 10.1017/s0954422423000124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
A model explaining the dietary-protein-driven post-natal skeletal muscle growth and protein turnover in the rat is updated, and the mechanisms involved are described, in this narrative review. Dietary protein controls both bone length and muscle growth, which are interrelated through mechanotransduction mechanisms with muscle growth induced both from stretching subsequent to bone length growth and from internal work against gravity. This induces satellite cell activation, myogenesis and remodelling of the extracellular matrix, establishing a growth capacity for myofibre length and cross-sectional area. Protein deposition within this capacity is enabled by adequate dietary protein and other key nutrients. After briefly reviewing the experimental animal origins of the growth model, key concepts and processes important for growth are reviewed. These include the growth in number and size of the myonuclear domain, satellite cell activity during post-natal development and the autocrine/paracrine action of IGF-1. Regulatory and signalling pathways reviewed include developmental mechanotransduction, signalling through the insulin/IGF-1-PI3K-Akt and the Ras-MAPK pathways in the myofibre and during mechanotransduction of satellite cells. Likely pathways activated by maximal-intensity muscle contractions are highlighted and the regulation of the capacity for protein synthesis in terms of ribosome assembly and the translational regulation of 5-TOPmRNA classes by mTORC1 and LARP1 are discussed. Evidence for and potential mechanisms by which volume limitation of muscle growth can occur which would limit protein deposition within the myofibre are reviewed. An understanding of how muscle growth is achieved allows better nutritional management of its growth in health and disease.
Collapse
Affiliation(s)
- D Joe Millward
- Department of Nutritional Sciences, School of Biosciences & Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
6
|
Cid-Bertomeu P, Vilaltella M, Martínez M, Mir M, Huerva V. Topical Insulin for Ocular Surface Disease. J Ocul Pharmacol Ther 2024; 40:204-214. [PMID: 38527183 DOI: 10.1089/jop.2024.0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024] Open
Abstract
Background: Insulin and insulin-like growth factor (IGF)-1 receptors are present in ocular tissues such as corneal epithelium, keratocytes, and conjunctival cells. Insulin plays a crucial role in the growth, differentiation, and proliferation of corneal epithelial cells, as well as in wound healing processes in various tissues. Purpose: This review explores the potential role of topical insulin in the treatment of ocular surface diseases. Specifically, it examines its impact on corneal nerve regeneration, sub-basal plexus corneal nerves, and its application in conditions like corneal epithelial defects, dry eye disease, and diabetic keratopathy. Methods: The review analyzes studies conducted over the past decade that have investigated the use of topical insulin in ocular surface diseases. It focuses on indications, drug preparation methods, side effects, efficacy outcomes, and variations in insulin concentrations and dosages used. Results: While off-label use of topical insulin has shown promising results in refractory corneal epithelial defects, its efficacy in dry eye disease is yet to be demonstrated. Variations in concentrations, dilutions, and dosing guidelines have been reported. However, limited data on ocular penetration, ocular toxicity, and systemic side effects pose challenges to its widespread utility. Conclusion: This review synthesizes findings from ocular investigations on topical insulin to assess its potential applicability in treating ocular surface and corneal diseases. By highlighting indications, preparation methods, side effects, and efficacy outcomes, it aims to provide insights into the current status and future prospects of using topical insulin in ophthalmic practice.
Collapse
Affiliation(s)
- Pau Cid-Bertomeu
- Department of Ophthalmology, University Hospital Arnau de Vilanova, Lleida, Spain
| | - Magí Vilaltella
- Department of Ophthalmology, University Hospital Arnau de Vilanova, Lleida, Spain
- School of Medicine, University of Lleida, Lleida, Spain
| | - Mireia Martínez
- Department of Hospital Pharmacy, University Hospital Arnau de Vilanova, Lleida, Spain
| | - Marta Mir
- Department of Hospital Pharmacy, University Hospital Arnau de Vilanova, Lleida, Spain
| | - Valentín Huerva
- Department of Ophthalmology, University Hospital Arnau de Vilanova, Lleida, Spain
- School of Medicine, University of Lleida, Lleida, Spain
| |
Collapse
|
7
|
Hayes E, Winston N, Stocco C. Molecular crosstalk between insulin-like growth factors and follicle-stimulating hormone in the regulation of granulosa cell function. Reprod Med Biol 2024; 23:e12575. [PMID: 38571513 PMCID: PMC10988955 DOI: 10.1002/rmb2.12575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/11/2024] [Accepted: 03/20/2024] [Indexed: 04/05/2024] Open
Abstract
Background The last phase of folliculogenesis is driven by follicle-stimulating hormone (FSH) and locally produced insulin-like growth factors (IGFs), both essential for forming preovulatory follicles. Methods This review discusses the molecular crosstalk of the FSH and IGF signaling pathways in regulating follicular granulosa cells (GCs) during the antral-to-preovulatory phase. Main findings IGFs were considered co-gonadotropins since they amplify FSH actions in GCs. However, this view is not compatible with data showing that FSH requires IGFs to stimulate GCs, that FSH renders GCs sensitive to IGFs, and that FSH signaling interacts with factors downstream of AKT to stimulate GCs. New evidence suggests that FSH and IGF signaling pathways intersect at several levels to regulate gene expression and GC function. Conclusion FSH and locally produced IGFs form a positive feedback loop essential for preovulatory follicle formation in all species. Understanding the mechanisms by which FSH and IGFs interact to control GC function will help design new interventions to optimize follicle maturation, perfect treatment of ovulatory defects, improve in vitro fertilization, and develop new contraceptive approaches.
Collapse
Affiliation(s)
- Emily Hayes
- Department of Physiology and BiophysicsUniversity of Illinois Chicago College of MedicineChicagoIllinoisUSA
| | - Nicola Winston
- Department of Obstetrics and GynecologyUniversity of Illinois Chicago College of MedicineChicagoIllinoisUSA
| | - Carlos Stocco
- Department of Physiology and BiophysicsUniversity of Illinois Chicago College of MedicineChicagoIllinoisUSA
- Department of Obstetrics and GynecologyUniversity of Illinois Chicago College of MedicineChicagoIllinoisUSA
| |
Collapse
|
8
|
Chen K, Dou X, Eum JH, Harrison RE, Brown MR, Strand MR. Insulin-like peptides and ovary ecdysteroidogenic hormone differentially stimulate physiological processes regulating egg formation in the mosquito Aedes aegypti. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 163:104028. [PMID: 37913852 PMCID: PMC10842226 DOI: 10.1016/j.ibmb.2023.104028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 10/12/2023] [Accepted: 10/23/2023] [Indexed: 11/03/2023]
Abstract
Mosquitoes including Aedes aegypti are human disease vectors because females must blood feed to produce and lay eggs. Blood feeding triggers insulin-insulin growth factor signaling (IIS) which regulates several physiological processes required for egg development. A. aegypti encodes 8 insulin-like peptides (ILPs) and one insulin-like receptor (IR) plus ovary ecdysteroidogenic hormone (OEH) that also activates IIS through the OEH receptor (OEHR). In this study, we assessed the expression of A. aegypti ILPs and OEH during a gonadotrophic cycle and produced each that were functionally characterized to further understand their roles in regulating egg formation. All A. aegypti ILPs and OEH were expressed during a gonadotrophic cycle. Five ILPs (1, 3, 4, 7, 8) and OEH were specifically expressed in the head, while antibodies to ILP3 and OEH indicated each was released after blood feeding from ventricular axons that terminate on the anterior midgut. A subset of ILP family members and OEH stimulated nutrient storage in previtellogenic females before blood feeding, whereas most IIS-dependent processes after blood feeding were activated by one or more of the brain-specific ILPs and/or OEH. ILPs and OEH with different biological activities also exhibited differences in IIS as measured by phosphorylation of the IR, phosphoinositide 3-kinase/Akt kinase (AKT) and mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK). Altogether, our results provide the first results that compare the functional activities of all ILP family members and OEH produced by an insect.
Collapse
Affiliation(s)
- Kangkang Chen
- Department of Entomology, University of Georgia, Athens, GA, USA
| | - Xiaoyi Dou
- Department of Entomology, University of Georgia, Athens, GA, USA
| | - Jai Hoon Eum
- Department of Entomology, University of Georgia, Athens, GA, USA
| | - Ruby E Harrison
- Department of Entomology, University of Georgia, Athens, GA, USA
| | - Mark R Brown
- Department of Entomology, University of Georgia, Athens, GA, USA.
| | - Michael R Strand
- Department of Entomology, University of Georgia, Athens, GA, USA.
| |
Collapse
|
9
|
Viola CM, Frittmann O, Jenkins HT, Shafi T, De Meyts P, Brzozowski AM. Structural conservation of insulin/IGF signalling axis at the insulin receptors level in Drosophila and humans. Nat Commun 2023; 14:6271. [PMID: 37805602 PMCID: PMC10560217 DOI: 10.1038/s41467-023-41862-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 09/21/2023] [Indexed: 10/09/2023] Open
Abstract
The insulin-related hormones regulate key life processes in Metazoa, from metabolism to growth, lifespan and aging, through an evolutionarily conserved insulin signalling axis (IIS). In humans the IIS axis is controlled by insulin, two insulin-like growth factors, two isoforms of the insulin receptor (hIR-A and -B), and its homologous IGF-1R. In Drosophila, this signalling engages seven insulin-like hormones (DILP1-7) and a single receptor (dmIR). This report describes the cryoEM structure of the dmIR ectodomain:DILP5 complex, revealing high structural homology between dmIR and hIR. The excess of DILP5 yields dmIR complex in an asymmetric 'T' conformation, similar to that observed in some complexes of human IRs. However, dmIR binds three DILP5 molecules in a distinct arrangement, showing also dmIR-specific features. This work adds structural support to evolutionary conservation of the IIS axis at the IR level, and also underpins a better understanding of an important model organism.
Collapse
Affiliation(s)
- Cristina M Viola
- York Structural Biology Laboratory, Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
- Department of Biology, University of York, Heslington, York, YO10 5DD, UK
| | - Orsolya Frittmann
- York Structural Biology Laboratory, Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
- Department of Haematology, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, Netherlands
| | - Huw T Jenkins
- York Structural Biology Laboratory, Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
| | - Talha Shafi
- York Structural Biology Laboratory, Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
| | - Pierre De Meyts
- Department of Cell Signalling, de Duve Institute, B-1200, Brussels, Belgium
- Department of Cell Therapy Research, Novo Nordisk A/S, DK-2670, Maaloev, Denmark
| | - Andrzej M Brzozowski
- York Structural Biology Laboratory, Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK.
| |
Collapse
|
10
|
Abstract
The insulin receptor (IR) is a type II receptor tyrosine kinase that plays essential roles in metabolism, growth, and proliferation. Dysregulation of IR signaling is linked to many human diseases, such as diabetes and cancers. The resolution revolution in cryo-electron microscopy has led to the determination of several structures of IR with different numbers of bound insulin molecules in recent years, which have tremendously improved our understanding of how IR is activated by insulin. Here, we review the insulin-induced activation mechanism of IR, including (a) the detailed binding modes and functions of insulin at site 1 and site 2 and (b) the insulin-induced structural transitions that are required for IR activation. We highlight several other key aspects of the activation and regulation of IR signaling and discuss the remaining gaps in our understanding of the IR activation mechanism and potential avenues of future research.
Collapse
Affiliation(s)
- Eunhee Choi
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA;
| | - Xiao-Chen Bai
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, USA;
| |
Collapse
|
11
|
Abstract
Insulin is a peptide hormone essential for maintaining normal blood glucose levels. Individuals unable to secrete sufficient insulin or not able to respond properly to insulin develop diabetes. Since the discovery of insulin its structure and function has been intensively studied with the aim to develop effective diabetes treatments. The three-dimensional crystal structure of this 51 amino acid peptide paved the way for discoveries, outlined in this review, of determinants important for receptor binding and hormone stability that have been instrumental in development of insulin analogs used in the clinic today. Important for the future development of effective diabetes treatments will be a detailed understanding of the insulin receptor structure and function. Determination of the three-dimensional structure of the insulin receptor, a receptor tyrosine kinase, proved challenging but with the recent advent of high-resolution cryo-electron microscopy significant progress has been made. There are now >40 structures of the insulin:insulin receptor complex deposited in the Protein Data Bank. From these structures we have a detailed picture of how insulin binds and activates the receptor. Still lacking are details of the initial binding events and the exact sequence of structural changes within the receptor and insulin. In this review, the focus will be on the most recent structural studies of insulin:insulin receptor complexes and how they have contributed to the current understanding of insulin receptor activation and signaling outcome. Molecular mechanisms underlying insulin receptor signaling bias emerging from the latest structures are described.
Collapse
Affiliation(s)
- Briony E Forbes
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia.
| |
Collapse
|
12
|
Viola CM, Frittmann O, Jenkins HT, Shafi T, Meyts PD, Brzozowski AM. Structural Conservation of Insulin/IGF Signalling Axis at the Insulin Receptors Level in Drosophilaand humans.. [DOI: 10.1101/2023.02.17.528932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
ABSTRACTThe insulin-related hormones regulate key life processes in Metazoa, from metabolism to growth, lifespan and aging, through an evolutionarily conserved insulin signalling axis (IIS). In humans the IIS axis is controlled by insulin, two insulin-like growth factors, two isoforms of the insulin receptor (hIR-A and -B), and its homologous IGF-1R. InDrosophila, this signalling engages seven insulin-like hormones (DILP1-7) and a single receptor (dmIR). This report describes the first cryoEM structure of the dmIR ectodomain:DILP5 complex, revealing high structural homology between dmIR and hIR. The excess of DILP5 yields dmIR complex in an asymmetric ‘T’ conformation, similar to that observed in some complexes of human IRs. However, dmIR binds three DILP5 molecules in a hitherto-unseen arrangement, showing also dmIR-specific features. This work adds structural support to evolutionary conservation of the IIS axis at the IRs levels, underpinning also a better understanding of an important model organism.
Collapse
|
13
|
Wang Y, Song X, Wang Y, Wang N. Specific interaction of insulin receptor and GLP-1 receptor mediates crosstalk between their signaling. Biochem Biophys Res Commun 2022; 636:31-39. [DOI: 10.1016/j.bbrc.2022.10.094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 10/27/2022] [Indexed: 11/02/2022]
|
14
|
Zhang X, Zhu X, Bi X, Huang J, Zhou L. The Insulin Receptor: An Important Target for the Development of Novel Medicines and Pesticides. Int J Mol Sci 2022; 23:7793. [PMID: 35887136 PMCID: PMC9325136 DOI: 10.3390/ijms23147793] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 02/04/2023] Open
Abstract
The insulin receptor (IR) is a transmembrane protein that is activated by ligands in insulin signaling pathways. The IR has been considered as a novel therapeutic target for clinical intervention, considering the overexpression of its protein and A-isoform in multiple cancers, Alzheimer's disease, and Type 2 diabetes mellitus in humans. Meanwhile, it may also serve as a potential target in pest management due to its multiple physiological influences in insects. In this review, we provide an overview of the structural and molecular biology of the IR, functions of IRs in humans and insects, physiological and nonpeptide small molecule modulators of the IR, and the regulating mechanisms of the IR. Xenobiotic compounds and the corresponding insecticidal chemicals functioning on the IR are also discussed. This review is expected to provide useful information for a better understanding of human IR-related diseases, as well as to facilitate the development of novel small-molecule activators and inhibitors of the IR for use as medicines or pesticides.
Collapse
Affiliation(s)
| | | | | | - Jiguang Huang
- Key Laboratory of Natural Pesticides & Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (X.Z.); (X.B.)
| | - Lijuan Zhou
- Key Laboratory of Natural Pesticides & Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (X.Z.); (X.B.)
| |
Collapse
|
15
|
Ballester-Rosado CJ, Le JT, Lam TT, Mohila CA, Lam S, Anderson AE, Frost JD, Swann JW. A Role for Insulin-like Growth Factor 1 in the Generation of Epileptic Spasms in a murine model. Ann Neurol 2022; 92:45-60. [PMID: 35467038 PMCID: PMC9233100 DOI: 10.1002/ana.26383] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 04/21/2022] [Accepted: 04/23/2022] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Infantile spasms are associated with a wide variety of clinical conditions, including perinatal brain injuries. We have created a model in which prolonged infusion of tetrodotoxin (TTX) into the neocortex, beginning in infancy, produces a localized lesion and reproduces the behavioral spasms, electroencephalogram (EEG) abnormalities, and drug responsiveness seen clinically. Here, we undertook experiments to explore the possibility that the growth factor IGF-1 plays a role in generating epileptic spasms. METHODS We combined long-term video EEG recordings with quantitative immunohistochemical and biochemical analyses to unravel IGF-1's role in spasm generation. Immunohistochemistry was undertaken in surgically resected tissue from infantile spasms patients. We used viral injections in neonatal conditional IGF-1R knock-out mice to show that an IGF-1-derived tripeptide (1-3)IGF-1, acts through the IGF-1 receptor to abolish spasms. RESULTS Immunohistochemical methods revealed widespread loss of IGF-1 from cortical neurons, but an increase in IGF-1 in the reactive astrocytes in the TTX-induced lesion. Very similar changes were observed in the neocortex from patients with spasms. In animals, we observed reduced signaling through the IGF-1 growth pathways in areas remote from the lesion. To show the reduction in IGF-1 expression plays a role in spasm generation, epileptic rats were treated with (1-3)IGF-1. We provide 3 lines of evidence that (1-3)IGF-1 activates the IGF-1 signaling pathway by acting through the receptor for IGF-1. Treatment with (1-3)IGF-1 abolished spasms and hypsarrhythmia-like activity in the majority of animals. INTERPRETATION Results implicate IGF-1 in the pathogenesis of infantile spasms and IGF-1 analogues as potential novel therapies for this neurodevelopmental disorder. ANN NEUROL 2022;92:45-60.
Collapse
Affiliation(s)
- Carlos J. Ballester-Rosado
- The Cain Foundation Laboratories, the Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - John T. Le
- The Cain Foundation Laboratories, the Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Trang T. Lam
- The Cain Foundation Laboratories, the Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Carrie A. Mohila
- Department of Pathology and Immunology, Baylor College of Medicine
- Department of Pathology, Texas Children’s Hospital, Houston, Texas, USA
| | - Sandi Lam
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA
| | - Anne E. Anderson
- The Cain Foundation Laboratories, the Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
- Department of Neurology, Baylor College of Medicine, Houston, Texas, USA
| | - James D. Frost
- Department of Neurology, Baylor College of Medicine, Houston, Texas, USA
| | - John W. Swann
- The Cain Foundation Laboratories, the Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
16
|
Shalitin S, Gat-Yablonski G. Associations of Obesity with Linear Growth and Puberty. Horm Res Paediatr 2022; 95:120-136. [PMID: 34130293 DOI: 10.1159/000516171] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 03/27/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The prevalence of obesity in childhood has increased dramatically in recent decades with increased risk of developing cardiometabolic and other comorbidities. Childhood adiposity may also influence processes of growth and puberty. SUMMARY Growth patterns of obesity during childhood have been shown to be associated with increased linear growth in early childhood, leading to accelerated epiphyseal growth plate (EGP) maturation. Several hormones secreted by the adipose tissue may affect linear growth in the context of obesity, both via the growth hormone IGF-1 axis and via a direct effect on the EGP. The observation that children with obesity tend to mature earlier than lean children has led to the assumption that the degree of body fatness may trigger the neuroendocrine events that lead to pubertal onset. The most probable link between obesity and puberty is leptin and its interaction with the kisspeptin system, which is an important regulator of puberty. However, peripheral action of adipose tissue could also be involved in changes in the onset of puberty. In addition, nutritional factors, epigenetics, and endocrine-disrupting chemicals are potential mediators linking pubertal onset to obesity. In this review, we focused on interactions of obesity with linear growth and pubertal processes, based on basic research and clinical data in humans. KEY MESSAGE Children with obesity are subject to accelerated linear growth with risk of impaired adult height and early puberty, with its psychological consequences. The data highlight another important objective in combatting childhood obesity, for the prevention of abnormal growth and pubertal patterns.
Collapse
Affiliation(s)
- Shlomit Shalitin
- National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Galia Gat-Yablonski
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Felsenstein Medical Research Center, Petach Tikva, Israel
| |
Collapse
|
17
|
Li H, Zhang J, Shi Y, Zhao G, Xu H, Cai M, Gao J, Wang H. Mechanism of INSR clustering with insulin activation and resistance revealed by super-resolution imaging. NANOSCALE 2022; 14:7747-7755. [PMID: 35579582 DOI: 10.1039/d2nr01051h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Insulin receptor (INSR) is a key protein in the INSR signaling pathway and plays a critical role in biological processes, especially in the regulation of glucose homeostasis. Many metabolic diseases are often accompanied by abnormal INSR signaling. However, the specific effector mechanisms regulating insulin resistance and the distribution patterns of INSR during cell membrane activation remain unclear. Here, we investigated the changes in the distribution of INSR during activation using super-resolution imaging. By observing the connection between INSR activation and its distribution, we found that insulin resistance inhibits its receptor clustering. More importantly, we found that INSR has a highly co-localized relationship with the skeletal protein βII-spectrin. Specific knockout of βII-spectrin inhibited the interaction of INSR with GLUT4 and affected the normal metabolism of glucose. Our work elucidates the effects of insulin activation and insulin resistance on INSR distribution and reveals a potential relationship between INSR and cytoskeleton at the single molecule level, which promotes a deeper understanding of the roles associated with insulin signaling and insulin resistance.
Collapse
Affiliation(s)
- Hongru Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.
- University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Jinrui Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.
| | - Yan Shi
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.
| | - Guanfang Zhao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.
- University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Haijiao Xu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.
| | - Mingjun Cai
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.
- University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Jing Gao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.
- University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qing dao National Laboratory for Marine Science and Technology, Wenhai Road, Aoshanwei, Jimo, Qingdao, Shandong 266237, China
| |
Collapse
|
18
|
Kamoshita K, Tsugane H, Ishii KA, Takayama H, Yao X, Abuduwaili H, Tanida R, Taniguchi Y, Oo HK, Gafiyatullina G, Kaneko S, Matsugo S, Takamura T. Lauric acid impairs insulin-induced Akt phosphorylation by upregulating SELENOP expression via HNF4α induction. Am J Physiol Endocrinol Metab 2022; 322:E556-E568. [PMID: 35499234 DOI: 10.1152/ajpendo.00163.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 04/06/2022] [Accepted: 04/25/2022] [Indexed: 11/22/2022]
Abstract
Selenoprotein P (SeP; encoded by SELENOP in humans, Selenop in rodents) is a hepatokine that is upregulated in the liver of humans with type 2 diabetes. Excess SeP contributes to the onset of insulin resistance and various type 2 diabetes-related complications. We have previously reported that the long-chain saturated fatty acid, palmitic acid, upregulates Selenop expression, whereas the polyunsaturated fatty acids (PUFAs) downregulate it in hepatocytes. However, the effect of medium-chain fatty acids (MCFAs) on Selenop is unknown. Here we report novel mechanisms that underlie the lauric acid-mediated Selenop gene regulation in hepatocytes. Lauric acid upregulated Selenop expression in Hepa1-6 hepatocytes and mice liver. A luciferase promoter assay and computational analysis of transcription factor-binding sites identified the hepatic nuclear factor 4α (HNF4α) binding site in the SELENOP promoter. A chromatin immunoprecipitation (ChIP) assay showed that lauric acid increased the binding of HNF4α to the SELENOP promoter. The knockdown of Hnf4α using siRNA canceled the upregulation of lauric acid-induced Selenop. Thus, the lauric acid-induced impairment of Akt phosphorylation brought about by insulin was rescued by the knockdown of either Hnf4α or Selenop. These results provide new insights into the regulation of SeP by fatty acids and suggest that SeP may mediate MCFA-induced hepatic insulin signal reduction.
Collapse
Affiliation(s)
- Kyoko Kamoshita
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| | - Hirohiko Tsugane
- Institute of Science and Engineering, Faculty of Natural System, Kanazawa University, Kanazawa, Japan
| | - Kiyo-Aki Ishii
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
- Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hiroaki Takayama
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
- Department of System Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
- Life Sciences Division, Engineering and Technology Department, Kanazawa University, Kanazawa, Japan
| | - Xingyu Yao
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| | - Halimulati Abuduwaili
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| | - Ryota Tanida
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| | - Yasumasa Taniguchi
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| | - Hein Ko Oo
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| | - Guzel Gafiyatullina
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| | - Shuichi Kaneko
- Department of System Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Seiichi Matsugo
- Institute of Science and Engineering, Faculty of Natural System, Kanazawa University, Kanazawa, Japan
| | - Toshinari Takamura
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| |
Collapse
|
19
|
Hutfilz C. Endocrine Regulation of Lifespan in Insect Diapause. Front Physiol 2022; 13:825057. [PMID: 35242054 PMCID: PMC8886022 DOI: 10.3389/fphys.2022.825057] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/25/2022] [Indexed: 01/27/2023] Open
Abstract
Diapause is a physiological adaptation to conditions that are unfavorable for growth or reproduction. During diapause, animals become long-lived, stress-resistant, developmentally static, and non-reproductive, in the case of diapausing adults. Diapause has been observed at all developmental stages in both vertebrates and invertebrates. In adults, diapause traits weaken into adaptations such as hibernation, estivation, dormancy, or torpor, which represent evolutionarily diverse versions of the traditional diapause traits. These traits are regulated through modifications of the endocrine program guiding development. In insects, this typically includes changes in molting hormones, as well as metabolic signals that limit growth while skewing the organism's energetic demands toward conservation. While much work has been done to characterize these modifications, the interactions between hormones and their downstream consequences are incompletely understood. The current state of diapause endocrinology is reviewed here to highlight the relevance of diapause beyond its use as a model to study seasonality and development. Specifically, insect diapause is an emerging model to study mechanisms that determine lifespan. The induction of diapause represents a dramatic change in the normal progression of age. Hormones such as juvenile hormone, 20-hydroxyecdysone, and prothoracicotropic hormone are well-known to modulate this plasticity. The induction of diapause-and by extension, the cessation of normal aging-is coordinated by interactions between these pathways. However, research directly connecting diapause endocrinology to the biology of aging is lacking. This review explores connections between diapause and aging through the perspective of endocrine signaling. The current state of research in both fields suggests appreciable overlap that will greatly contribute to our understanding of diapause and lifespan determination.
Collapse
|
20
|
Li J, Wu J, Hall C, Bai XC, Choi E. Molecular basis for the role of disulfide-linked αCTs in the activation of insulin-like growth factor 1 receptor and insulin receptor. eLife 2022; 11:81286. [PMID: 36413010 PMCID: PMC9731570 DOI: 10.7554/elife.81286] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022] Open
Abstract
The insulin receptor (IR) and insulin-like growth factor 1 receptor (IGF1R) control metabolic homeostasis and cell growth and proliferation. The IR and IGF1R form similar disulfide bonds linked homodimers in the apo-state; however, their ligand binding properties and the structures in the active state differ substantially. It has been proposed that the disulfide-linked C-terminal segment of α-chain (αCTs) of the IR and IGF1R control the cooperativity of ligand binding and regulate the receptor activation. Nevertheless, the molecular basis for the roles of disulfide-linked αCTs in IR and IGF1R activation are still unclear. Here, we report the cryo-EM structures of full-length mouse IGF1R/IGF1 and IR/insulin complexes with modified αCTs that have increased flexibility. Unlike the Γ-shaped asymmetric IGF1R dimer with a single IGF1 bound, the IGF1R with the enhanced flexibility of αCTs can form a T-shaped symmetric dimer with two IGF1s bound. Meanwhile, the IR with non-covalently linked αCTs predominantly adopts an asymmetric conformation with four insulins bound, which is distinct from the T-shaped symmetric IR. Using cell-based experiments, we further showed that both IGF1R and IR with the modified αCTs cannot activate the downstream signaling potently. Collectively, our studies demonstrate that the certain structural rigidity of disulfide-linked αCTs is critical for optimal IR and IGF1R signaling activation.
Collapse
Affiliation(s)
- Jie Li
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Jiayi Wu
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia UniversityNew YorkUnited States
| | - Catherine Hall
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia UniversityNew YorkUnited States
| | - Xiao-chen Bai
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States,Department of Cell Biology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Eunhee Choi
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia UniversityNew YorkUnited States
| |
Collapse
|
21
|
Brierley GV, Semple RK. Insulin at 100 years - is rebalancing its action key to fighting obesity-related disease? Dis Model Mech 2021; 14:273551. [PMID: 34841432 PMCID: PMC8649170 DOI: 10.1242/dmm.049340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
One hundred years ago, insulin was purified and administered to people with diabetes to lower blood glucose, suppress ketogenesis and save lives. A century later, insulin resistance (IR) lies at the heart of the obesity-related disease pandemic. Multiple observations attest that IR syndrome is an amalgamation of gain and loss of insulin action, suggesting that IR is a misnomer. This misapprehension is reinforced by shortcomings in common model systems and is particularly pronounced for the tissue growth disorders associated with IR. It is necessary to move away from conceptualisation of IR as a pure state of impaired insulin action and to appreciate that, in the long term, insulin can harm as well as cure. The mixed state of gain and loss of insulin action, and its relationship to perturbed insulin-like growth factor (IGF) action, should be interrogated more fully in models recapitulating human disease. Only then may the potential of rebalancing insulin action, rather than simply increasing global insulin signalling, finally be appreciated.
Collapse
Affiliation(s)
- Gemma V Brierley
- Biomedical Research Group, School of Life Sciences, Anglia Ruskin University, Cambridge CB1 1PT, UK.,The University of Cambridge Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Robert K Semple
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| |
Collapse
|
22
|
Pandey M, Cuddihy G, Gordon JA, Cox ME, Wasan KM. Inhibition of Scavenger Receptor Class B Type 1 (SR-B1) Expression and Activity as a Potential Novel Target to Disrupt Cholesterol Availability in Castration-Resistant Prostate Cancer. Pharmaceutics 2021; 13:1509. [PMID: 34575583 PMCID: PMC8467449 DOI: 10.3390/pharmaceutics13091509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023] Open
Abstract
There have been several studies that have linked elevated scavenger receptor class b type 1 (SR-B1) expression and activity to the development and progression of castration-resistant prostate cancer (CRPC). SR-B1 facilitates the influx of cholesterol to the cell from lipoproteins in systemic circulation. This influx of cholesterol may be important for many cellular functions, including the synthesis of androgens. Castration-resistant prostate cancer tumors can synthesize androgens de novo to supplement the loss of exogenous sources often induced by androgen deprivation therapy. Silencing of SR-B1 may impact the ability of prostate cancer cells, particularly those of the castration-resistant state, to maintain the intracellular supply of androgens by removing a supply of cholesterol. SR-B1 expression is elevated in CRPC models and has been linked to poor survival of patients. The overarching belief has been that cholesterol modulation, through either synthesis or uptake inhibition, will impact essential signaling processes, impeding the proliferation of prostate cancer. The reduction in cellular cholesterol availability can impede prostate cancer proliferation through both decreased steroid synthesis and steroid-independent mechanisms, providing a potential therapeutic target for the treatment of prostate cancer. In this article, we discuss and highlight the work on SR-B1 as a potential novel drug target for CRPC management.
Collapse
Affiliation(s)
- Mitali Pandey
- Department of Urological Sciences, Faculty of Medicine, University of British Columbia, Vancouver Prostate Centre, Vancouver, BC V6T 1Z3, Canada; (M.P.); (M.E.C.)
| | - Grace Cuddihy
- College of Pharmacy and Nutrition, University of Saskatchewan, 104 Clinic Place, Saskatoon, SK S7N 2Z4, Canada;
| | - Jacob A. Gordon
- Oncology Bioscience, Oncology R&D, AstraZeneca, Boston, MA 02451, USA;
| | - Michael E. Cox
- Department of Urological Sciences, Faculty of Medicine, University of British Columbia, Vancouver Prostate Centre, Vancouver, BC V6T 1Z3, Canada; (M.P.); (M.E.C.)
| | - Kishor M. Wasan
- Department of Urological Sciences, Faculty of Medicine, University of British Columbia, Vancouver Prostate Centre, Vancouver, BC V6T 1Z3, Canada; (M.P.); (M.E.C.)
| |
Collapse
|
23
|
Evolution and medicine - The central role of anatomy. Ann Anat 2021; 239:151809. [PMID: 34324995 DOI: 10.1016/j.aanat.2021.151809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 06/10/2021] [Accepted: 07/13/2021] [Indexed: 11/24/2022]
Abstract
In medicine, there is an increasing number of publications that deal with or at least consider an evolutionary background. In zoology or comparative anatomy, work on evolutionary developments is taking on an ever-greater role in parallel. The pre-clinical (or pre-medical) phase in medical studies would be able to form a bridge between these related and yet so distant subjects but is currently completely evolution-free. This means that there is no consideration of the evolution of the healthy human being as a prerequisite for a systematic study of the evolutionary background in medicine. In this work the view is expressed that anatomy should be given a central, framework-giving and integrating role, which should urgently be actively pursued.
Collapse
|
24
|
Giglione E, Lapolla R, Cianfarani S, Faienza MF, Fintini D, Weber G, Delvecchio M, Valerio G. Linear growth and puberty in childhood obesity: what is new? Minerva Pediatr (Torino) 2021; 73:563-571. [PMID: 34309346 DOI: 10.23736/s2724-5276.21.06543-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Pediatric obesity is a growing and alarming global health problem and represents an important determinant of morbidity. Since nutrition plays an important role in regulating growth and development, the excess weight gain related to overnutrition can affect growth patterns, bone maturation and pubertal development. The purpose of this review is to summarize the current knowledge about the effect of primary obesity on linear growth and pubertal development in children and adolescents. Evidences about regulatory hormones and adipokines that may be involved in the physiology of childhood growth in the context of obesity were also discussed. The most recent literature confirms previous studies indicating that linear growth is accelerated (mainly due to longer trunks rather than longer legs) and bone age is advanced in prepubertal children with obesity, while there is a reduction of pubertal height gain and attainment of normal adult height. Conflicting results are reported on the timing of puberty, specifically in boys. Indeed, previous studies suggested earlier onset of puberty in obese girls and overweight boys, and a delayed puberty in obese boys. Conversely, the most recent studies show more consistently an earlier onset and completion of pubertal development also in boys with obesity. Considering the false belief of health associated with transient taller stature in children and the adverse outcomes related to early puberty, interventions on diet and physical activity are urgently needed to tackle the epidemics of childhood obesity in public health and clinical setting.
Collapse
Affiliation(s)
| | - Rosa Lapolla
- Dipartimento Materno-Infantile, AOR San Carlo, Potenza, Italy
| | - Stefano Cianfarani
- Dipartimento Pediatrico Universitario Ospedaliero, IRCCS Bambino Gesù Children's Hospital, Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,Department of Women's and Children's Health, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Maria F Faienza
- Department of Biomedical Sciences and Human Oncology, Pediatric Unit, University A. Moro, Bari, Italy
| | - Danilo Fintini
- Endocrinology Unit, Pediatric University Department, Bambino Gesù Children Hospital, Rome, Italy
| | - Giovanna Weber
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
| | - Maurizio Delvecchio
- Metabolic Disorders and Genetic Unit, Giovanni XXIII Children Hospital, Bari, Italy
| | - Giuliana Valerio
- Department of Movement Sciences and Wellbeing, University of Naples Parthenope, Naples, Italy -
| |
Collapse
|
25
|
Gram M, Ekström C, Holmqvist B, Carey G, Wang X, Vallius S, Hellström W, Ortenlöf N, Agyemang AA, Smith LEH, Hellström A, Mangili A, Barton N, Ley D. Insulin-Like Growth Factor 1 in the Preterm Rabbit Pup: Characterization of Cerebrovascular Maturation following Administration of Recombinant Human Insulin-Like Growth Factor 1/Insulin-Like Growth Factor 1-Binding Protein 3. Dev Neurosci 2021; 43:281-295. [PMID: 34218224 PMCID: PMC8623584 DOI: 10.1159/000516665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/28/2021] [Indexed: 11/19/2022] Open
Abstract
Following preterm birth, serum levels of insulin-like growth factor 1 (IGF-1) decrease compared to corresponding in utero levels. A recent clinical trial indicated that supplementation with recombinant human (rh) IGF-1/rhIGF-binding protein 3 (rhIGF-1/rhIGFBP-3) prevents severe intraventricular hemorrhage (IVH) in extremely preterm infants. In a preterm rabbit pup model, we characterized endogenous serum and hepatic IGF-1, along with brain distribution of IGF-1 and IGF-1 receptor (IGF1R). We then evaluated the effects of rhIGF-1/rhIGFBP-3 on gene expression of regulators of cerebrovascular maturation and structure. Similar to preterm infants, serum IGF-1 concentrations decreased rapidly after preterm birth in the rabbit pup. Administration of rhIGF-1/rhIGFBP-3 restored in utero serum levels but was rapidly eliminated. Immunolabeled IGF1R was widely distributed in multiple brain regions, displaying an abundant density in the choroid plexus and sub-ependymal germinal zones. Increased IGF-1 immunoreactivity, distributed as IGF1R, was detected 4 h after rhIGF-1/rhIGFBP-3 administration. The rhIGF-1/rhIGFBP-3 treatment led to upregulation of choroid plexus genes involved in vascular maturation and structure, with corresponding protein translation for most of these genes. The preterm rabbit pup model is well suited for evaluation of IGF-1-based prevention of IVH. Administration of rhIGF-1/rhIGFBP-3 affects cerebrovascular maturation, suggesting a role for it in preventing preterm IVH.
Collapse
Affiliation(s)
- Magnus Gram
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Lund, Sweden,
| | - Claes Ekström
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Lund, Sweden
| | | | - Galen Carey
- Takeda Pharmaceuticals, Boston, Massachusetts, USA
| | - Xiaoyang Wang
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Suvi Vallius
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Lund, Sweden
| | - William Hellström
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Niklas Ortenlöf
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Lund, Sweden
| | | | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ann Hellström
- Department of Clinical Neuroscience, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alexandra Mangili
- Global Clinical Development, Rare Metabolic Diseases, Shire, a Takeda Company, Zurich, Switzerland
| | | | - David Ley
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Lund, Sweden
| |
Collapse
|
26
|
Distinct signaling by insulin and IGF-1 receptors and their extra- and intracellular domains. Proc Natl Acad Sci U S A 2021; 118:2019474118. [PMID: 33879610 DOI: 10.1073/pnas.2019474118] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Insulin and insulin-like growth factor 1 (IGF-1) receptors share many downstream signaling pathways but have unique biological effects. To define the molecular signals contributing to these distinct activities, we performed global phosphoproteomics on cells expressing either insulin receptor (IR), IGF-1 receptor (IGF1R), or chimeric IR-IGF1R receptors. We show that IR preferentially stimulates phosphorylations associated with mammalian target of rapamycin complex 1 (mTORC1) and Akt pathways, whereas IGF1R preferentially stimulates phosphorylations on proteins associated with the Ras homolog family of guanosine triphosphate hydrolases (Rho GTPases), and cell cycle progression. There were also major differences in the phosphoproteome between cells expressing IR versus IGF1R in the unstimulated state, including phosphorylation of proteins involved in membrane trafficking, chromatin remodeling, and cell cycle. In cells expressing chimeric IR-IGF1R receptors, these differences in signaling could be mapped to contributions of both the extra- and intracellular domains of these receptors. Thus, despite their high homology, IR and IGF1R preferentially regulate distinct networks of phosphorylation in both the basal and stimulated states, allowing for the unique effects of these hormones on organismal function.
Collapse
|
27
|
Janssen JA, Smith TJ. Lessons Learned from Targeting IGF-I Receptor in Thyroid-Associated Ophthalmopathy. Cells 2021; 10:cells10020383. [PMID: 33673340 PMCID: PMC7917650 DOI: 10.3390/cells10020383] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/21/2022] Open
Abstract
Complex immunological mechanisms underlie the pathogenesis of thyroid-associated ophthalmopathy (TAO). Historical models of Graves’ disease and TAO have focused almost entirely on autoimmune reactivity directed against the thyrotropin receptor (TSHR). The insulin-like growth factor-I receptor (IGF-IR) has been proposed as a second participating antigen in TAO by virtue of its interactions with IGFs and anti-IGF-IR antibodies generated in Graves’ disease. Furthermore, the IGF-IR forms with TSHR a physical and functional complex which is involved in signaling downstream from both receptors. Inhibition of IGF-IR activity results in attenuation of signaling initiated at either receptor. Based on the aggregate of findings implicating IGF-IR in TAO, the receptor has become an attractive therapeutic target. Recently, teprotumumab, a human monoclonal antibody IGF-IR inhibitor was evaluated in two clinical trials of patients with moderate to severe, active TAO. Those studies revealed that teprotumumab was safe and highly effective in reducing disease activity and severity. Targeting IGF-IR with specific biologic agents may result in a paradigm shift in the therapy of TAO.
Collapse
Affiliation(s)
- Joseph A.M.J.L. Janssen
- Erasmus Medical Center, Department of Internal Medicine, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
- Correspondence: ; Tel.: +31-10-7040704
| | - Terry J. Smith
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, MI 48105, USA;
- Division of Metabolism, Department of Internal Medicine, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| |
Collapse
|
28
|
Beletskiy A, Chesnokova E, Bal N. Insulin-Like Growth Factor 2 As a Possible Neuroprotective Agent and Memory Enhancer-Its Comparative Expression, Processing and Signaling in Mammalian CNS. Int J Mol Sci 2021; 22:ijms22041849. [PMID: 33673334 PMCID: PMC7918606 DOI: 10.3390/ijms22041849] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/13/2022] Open
Abstract
A number of studies performed on rodents suggest that insulin-like growth factor 2 (IGF-2) or its analogs may possibly be used for treating some conditions like Alzheimer’s disease, Huntington’s disease, autistic spectrum disorders or aging-related cognitive impairment. Still, for translational research a comparative knowledge about the function of IGF-2 and related molecules in model organisms (rats and mice) and humans is necessary. There is a number of important differences in IGF-2 signaling between species. In the present review we emphasize species-specific patterns of IGF-2 expression in rodents, humans and some other mammals, using, among other sources, publicly available transcriptomic data. We provide a detailed description of Igf2 mRNA expression regulation and pre-pro-IGF-2 protein processing in different species. We also summarize the function of IGF-binding proteins. We describe three different receptors able to bind IGF-2 and discuss the role of IGF-2 signaling in learning and memory, as well as in neuroprotection. We hope that comprehensive understanding of similarities and differences in IGF-2 signaling between model organisms and humans will be useful for development of more effective medicines targeting IGF-2 receptors.
Collapse
|
29
|
Hengpratom T, Lowe GM, Eumkeb G. An insight into anti-adipogenic properties of an Oroxylum indicum (L.) Kurz extract. BMC Complement Med Ther 2020; 20:319. [PMID: 33081786 PMCID: PMC7576871 DOI: 10.1186/s12906-020-03111-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/08/2020] [Indexed: 11/23/2022] Open
Abstract
Background Oroxylum indicum fruit extract (OIE) has been reported to inhibit the development of adipocytes. However, the exact mechanism of its metabolic activity is not clearly defined. This study attempted to investigate whether OIE was involved in disrupting the cell cycle, glucose metabolism, and mitochondrial function in 3 T3-L1 cells. Methods The effect of the OIE on cell cycle progression was measured by flow cytometry along with observing the expression of the cycle regulator by immunoblotting. The effect of the OIE on glucose metabolism was investigated. The amount of glucose uptake (2-NBDG) influenced by insulin was determined as well as the protein tyrosine phosphorylation (PY20), and glucose transporter4 (GLUT4) expression was determined by immunoblotting assay. Mitochondria are also essential to metabolic processes. This study investigated mitochondrial activity using fluorescent lipophilic carbocyanine dye (JC-1) and mitochondria mass by MitoTracker Green (MTG) staining fluorescence dyes. Finally, cellular ATP concentration was measured using an ATP chemiluminescence assay. Results Treatment with OIE plus adipogenic stimulators for 24 h arrested cell cycle progression in the G2/M phase. Moreover, 200 μg/mL of OIE significantly diminished the expression of the insulin receptor (IR) and GLUT4 protein compared to the untreated-adipocytes (P < 0.05). The mitochondrial membrane potential (MMP) was significantly reduced (24 h) and increased (day 12) by OIE compared to untreated-adipocytes (P < 0.05). However, OIE maintained MMP and ATP at a similar level compared to the pre-adipocytes (day 12). Transmission electron microscope (TEM) results demonstrated that OIE could protect mitochondria deformation compared to the untreated-adipocytes. Conclusion These results suggest that the inhibitory effect of the OIE on adipogenesis may potentially inhibit the cell cycle and phosphorylation of IR, leading to a decrease in glucose uptake to the cells. The OIE also slows down the mitochondrial activity of the early phase of cell differentiation, which can also inhibit the development of fat cells. Supplementary information The online version contains supplementary material available at 10.1186/s12906-020-03111-2.
Collapse
Affiliation(s)
- Tanaporn Hengpratom
- School of Preclinic, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, 30000, Thailand
| | - Gordon M Lowe
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, L3 3AF, UK.
| | - Griangsak Eumkeb
- School of Preclinic, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, 30000, Thailand.
| |
Collapse
|
30
|
Pek NMQ, Liu KJ, Nichane M, Ang LT. Controversies Surrounding the Origin of Hepatocytes in Adult Livers and the in Vitro Generation or Propagation of Hepatocytes. Cell Mol Gastroenterol Hepatol 2020; 11:273-290. [PMID: 32992051 PMCID: PMC7695885 DOI: 10.1016/j.jcmgh.2020.09.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 12/21/2022]
Abstract
Epithelial cells in the liver (known as hepatocytes) are high-performance engines of myriad metabolic functions and versatile responders to liver injury. As hepatocytes metabolize amino acids, alcohol, drugs, and other substrates, they produce and are exposed to a milieu of toxins and harmful byproducts that can damage themselves. In the healthy liver, hepatocytes generally divide slowly. However, after liver injury, hepatocytes can ramp up proliferation to regenerate the liver. Yet, on extensive injury, regeneration falters, and liver failure ensues. It is therefore critical to understand the mechanisms underlying liver regeneration and, in particular, which liver cells are mobilized during liver maintenance and repair. Controversies continue to surround the very existence of hepatic stem cells and, if they exist, their spatial location, multipotency, degree of contribution to regeneration, ploidy, and susceptibility to tumorigenesis. This review discusses these controversies. Finally, we highlight how insights into hepatocyte regeneration and biology in vivo can inform in vitro studies to propagate primary hepatocytes with liver regeneration-associated signals and to generate hepatocytes de novo from pluripotent stem cells.
Collapse
Affiliation(s)
| | | | | | - Lay Teng Ang
- Correspondence Address correspondence to: Lay Teng Ang, PhD, Stanford Institute for Stem Cell Biology & Regenerative Medicine, Stanford-UC Berkeley Siebel Stem Cell Institute, Stanford University School of Medicine, Stanford, California 94305.
| |
Collapse
|
31
|
Holly JMP, Biernacka K, Perks CM. The role of insulin-like growth factors in the development of prostate cancer. Expert Rev Endocrinol Metab 2020; 15:237-250. [PMID: 32441162 DOI: 10.1080/17446651.2020.1764844] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/01/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Preclinical, clinical, and population studies have provided robust evidence for an important role for the insulin-like growth factor (IGF) system in the development of prostate cancer. AREAS COVERED An overview of the IGF system is provided. The evidence implicating the IGF system in the development of prostate cancer is summarized. The compelling evidence culminated in a number of clinical trials of agents targeting the system; the reasons for the failure of these trials are discussed. EXPERT OPINION Clinical trials of agents targeting the IGF system in prostate cancer were terminated due to limited objective clinical responses and are unlikely to be resumed unless a convincing predictive biomarker is identified that would enable the selection of likely responders. The aging population and increased screening will lead to greater diagnosis of prostate cancer. Although the vast majority will be indolent disease, the epidemics of obesity and diabetes will increase the proportion that progress to clinical disease. The increased population of worried men will result in more trials aimed to reduce the risk of disease progression; actual clinical endpoints will be challenging and the IGFs remain the best intermediate biomarkers to indicate a response that could alter the course of disease.
Collapse
Affiliation(s)
- Jeff M P Holly
- IGFs & Metabolic Endocrinology Group, Faculty of Health Sciences, School of Translational Health Science, University of Bristol, Southmead Hospital , Bristol, UK
| | - Kalina Biernacka
- IGFs & Metabolic Endocrinology Group, Faculty of Health Sciences, School of Translational Health Science, University of Bristol, Southmead Hospital , Bristol, UK
| | - Claire M Perks
- IGFs & Metabolic Endocrinology Group, Faculty of Health Sciences, School of Translational Health Science, University of Bristol, Southmead Hospital , Bristol, UK
| |
Collapse
|
32
|
Luo J, Ting CY, Li Y, McQueen P, Lin TY, Hsu CP, Lee CH. Antagonistic regulation by insulin-like peptide and activin ensures the elaboration of appropriate dendritic field sizes of amacrine neurons. eLife 2020; 9:50568. [PMID: 32175842 PMCID: PMC7075694 DOI: 10.7554/elife.50568] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 03/05/2020] [Indexed: 01/09/2023] Open
Abstract
Establishing appropriate sizes and shapes of dendritic arbors is critical for proper wiring of the central nervous system. Here we report that Insulin-like Peptide 2 (DILP2) locally activates transiently expressed insulin receptors in the central dendrites of Drosophila Dm8 amacrine neurons to positively regulate dendritic field elaboration. We found DILP2 was expressed in L5 lamina neurons, which have axonal terminals abutting Dm8 dendrites. Proper Dm8 dendrite morphogenesis and synapse formation required insulin signaling through TOR (target of rapamycin) and SREBP (sterol regulatory element-binding protein), acting in parallel with previously identified negative regulation by Activin signaling to provide robust control of Dm8 dendrite elaboration. A simulation of dendritic growth revealed trade-offs between dendritic field size and robustness when branching and terminating kinetic parameters were constant, but dynamic modulation of the parameters could mitigate these trade-offs. We suggest that antagonistic DILP2 and Activin signals from different afferents appropriately size Dm8 dendritic fields.
Collapse
Affiliation(s)
- Jiangnan Luo
- Section on Neuronal Connectivity, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Chun-Yuan Ting
- Section on Neuronal Connectivity, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Yan Li
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Philip McQueen
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, United States
| | - Tzu-Yang Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Chao-Ping Hsu
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan, Republic of China.,Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei, Taiwan, Republic of China
| | - Chi-Hon Lee
- Section on Neuronal Connectivity, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States.,Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan, Republic of China
| |
Collapse
|
33
|
Li Z, Xu K, Guo Y, Ping L, Gao Y, Qiu Y, Ni J, Liu Q, Wang Z. A high-fat diet reverses metabolic disorders and premature aging by modulating insulin and IGF1 signaling in SIRT6 knockout mice. Aging Cell 2020; 19:e13104. [PMID: 31967391 PMCID: PMC7059164 DOI: 10.1111/acel.13104] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 12/04/2019] [Accepted: 12/30/2019] [Indexed: 12/26/2022] Open
Abstract
Mammalian sirtuin 6 (SIRT6) is involved in the regulation of many essential processes, especially metabolic homeostasis. SIRT6 knockout mice undergo premature aging and die at age ~4 weeks. Severe glycometabolic disorders have been found in SIRT6 knockout mice, and whether a dietary intervention can rescue SIRT6 knockout mice remains unknown. In our study, we found that at the same calorie intake, a high‐fat diet dramatically increased the lifespan of SIRT6 knockout mice to 26 weeks (males) and 37 weeks (females), reversed multi‐organ atrophy, and reduced body weight, hypoglycemia, and premature aging. Furthermore, the high‐fat diet partially but significantly normalized the global gene expression profile in SIRT6 knockout mice. Regarding the mechanism, excessive glucose uptake and glycolysis induced by the SIRT6 deficiency were attenuated in skeletal muscle through inhibition of insulin and IGF1 signaling by the high‐fat diet. Similarly, fatty acids but not ketone bodies inhibited glucose uptake, glycolysis, and senescence in SIRT6 knockout fibroblasts, whereas PI3K inhibition antagonized the effects of a high‐fatty‐acid medium in vitro. Overall, the high‐fat diet dramatically reverses numerous consequences of SIRT6 deficiency through modulation of insulin and IGF1 signaling, providing a new basis for elucidation of SIRT6 and fatty‐acid functions and supporting novel therapeutic approaches against metabolic disorders and aging‐related diseases.
Collapse
Affiliation(s)
- Zhongchi Li
- Protein Science Key Laboratory of the Ministry of Education School of Pharmaceutical Sciences Tsinghua University Beijing China
| | - Kang Xu
- Protein Science Key Laboratory of the Ministry of Education School of Pharmaceutical Sciences Tsinghua University Beijing China
| | - Yannan Guo
- Protein Science Key Laboratory of the Ministry of Education School of Pharmaceutical Sciences Tsinghua University Beijing China
| | - Lu Ping
- 8‐year MD Program Peking Union Medical College Beijing China
| | - Yuqi Gao
- Protein Science Key Laboratory of the Ministry of Education School of Pharmaceutical Sciences Tsinghua University Beijing China
| | - Ying Qiu
- School of Medicine Tsinghua University Beijing China
| | - Jianquan Ni
- School of Medicine Tsinghua University Beijing China
| | - Qingfei Liu
- Protein Science Key Laboratory of the Ministry of Education School of Pharmaceutical Sciences Tsinghua University Beijing China
| | - Zhao Wang
- Protein Science Key Laboratory of the Ministry of Education School of Pharmaceutical Sciences Tsinghua University Beijing China
| |
Collapse
|
34
|
Zhang ZD, Ren H, Wang WX, Shen GY, Huang JJ, Zhan MQ, Tang JJ, Yu X, Zhang YZ, Liang D, Yang ZD, Jiang XB. IGF-1R/β-catenin signaling axis is involved in type 2 diabetic osteoporosis. J Zhejiang Univ Sci B 2020; 20:838-848. [PMID: 31489803 DOI: 10.1631/jzus.b1800648] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Insulin-like growth factor-1 receptor (IGF-1R) is involved in both glucose and bone metabolism. IGF-1R signaling regulates the canonical Wnt/β-catenin signaling pathway. In this study, we investigated whether the IGF-1R/ β-catenin signaling axis plays a role in the pathogenesis of diabetic osteoporosis (DOP). Serum from patients with or without DOP was collected to measure the IGF-1R level using enzyme-linked immunosorbent assay (ELISA). Rats were given streptozotocin following a four-week high-fat diet induction (DOP group), or received vehicle after the same period of a normal diet (control group). Dual energy X-ray absorption, a biomechanics test, and hematoxylin-eosin (HE) staining were performed to evaluate bone mass, bone strength, and histomorphology, respectively, in vertebrae. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting were performed to measure the total and phosphorylation levels of IGF-1R, glycogen synthase kinase-3β (GSK-3β), and β-catenin. The serum IGF-1R level was much higher in patients with DOP than in controls. DOP rats exhibited strikingly reduced bone mass and attenuated compression strength of the vertebrae compared with the control group. HE staining showed that the histomorphology of DOP vertebrae was seriously impaired, which manifested as decreased and thinned trabeculae and increased lipid droplets within trabeculae. PCR analysis demonstrated that IGF-1R mRNA expression was significantly up-regulated, and western blotting detection showed that phosphorylation levels of IGF-1R, GSK-3β, and β-catenin were enhanced in DOP rat vertebrae. Our results suggest that the IGF-1R/β-catenin signaling axis plays a role in the pathogenesis of DOP. This may contribute to development of the underlying therapeutic target for DOP.
Collapse
Affiliation(s)
- Zhi-Da Zhang
- The First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Hui Ren
- Department of Spinal Surgery, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Wei-Xi Wang
- The First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Geng-Yang Shen
- Department of Spinal Surgery, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jin-Jing Huang
- The First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Mei-Qi Zhan
- The First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jing-Jing Tang
- Department of Spinal Surgery, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiang Yu
- The First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yu-Zhuo Zhang
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - De Liang
- Department of Spinal Surgery, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zhi-Dong Yang
- Department of Spinal Surgery, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiao-Bing Jiang
- Department of Spinal Surgery, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| |
Collapse
|
35
|
Sekulovski N, Whorton AE, Shi M, Hayashi K, MacLean JA. Periovulatory insulin signaling is essential for ovulation, granulosa cell differentiation, and female fertility. FASEB J 2020; 34:2376-2391. [PMID: 31908002 PMCID: PMC7781071 DOI: 10.1096/fj.201901791r] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/20/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022]
Abstract
Recent studies have demonstrated an essential role for insulin signaling in folliculogenesis as conditional ablation of Igf1r in primary follicles elicits defective follicle-stimulating hormone responsiveness blocking development at the preantral stage. Thus the potential role of insulin action in the periovulatory window and in the corpus luteum is unknown. To examine this, we generated conditional Insr,Igf1r, and double receptor knockout mice driven by Pgr-Cre. These models escape the preantral follicle block and in response to superovulatory gonadotropins exhibit normal distribution of ovarian follicles and corpora lutea. However, single ablation of Igf1r leads to subfertility and mice lacking both receptors are infertile. Double knockout mice have impaired oocyte development and ovulation. While some oocytes are released and fertilized, subsequent embryo development is retarded, and the embryos potentially fail to thrive due to lack of luteal support. In support of this, we found reduced expression of key enzymes in the steroid synthesis pathway and reduced serum progesterone. In addition to metabolic and steroidogenic pathways, RNA-sequencing analysis revealed transcription factor-3 as an important transcription factor downstream of insulin signaling. Collectively, these results highlight the importance of growth factors of the insulin family during two distinct windows of follicular development, ovulation, and luteinization.
Collapse
Affiliation(s)
- Nikola Sekulovski
- Department of Physiology, Southern Illinois University School of Medicine, Life Science III, Carbondale, IL, USA
| | - Allison E Whorton
- Department of Physiology, Southern Illinois University School of Medicine, Life Science III, Carbondale, IL, USA
| | - Mingxin Shi
- Department of Physiology, Southern Illinois University School of Medicine, Life Science III, Carbondale, IL, USA
| | - Kanako Hayashi
- Department of Physiology, Southern Illinois University School of Medicine, Life Science III, Carbondale, IL, USA
| | - James A MacLean
- Department of Physiology, Southern Illinois University School of Medicine, Life Science III, Carbondale, IL, USA
| |
Collapse
|
36
|
Minnetti M, Caiulo S, Ferrigno R, Baldini-Ferroli B, Bottaro G, Gianfrilli D, Sbardella E, De Martino MC, Savage MO. Abnormal linear growth in paediatric adrenal diseases: Pathogenesis, prevalence and management. Clin Endocrinol (Oxf) 2020; 92:98-108. [PMID: 31747461 DOI: 10.1111/cen.14131] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/10/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022]
Abstract
Abnormal adrenal function can interfere with linear growth, potentially causing either acceleration or impairment of growth in paediatric patients. These abnormalities can be caused by direct effects of adrenal hormones, particularly glucocorticoids and sex steroids, or be mediated by indirect mechanisms such as the disturbance of the growth hormone-insulin-like growth factor-1 axis and aromatization of androgens to oestrogens. The early diagnosis and optimal treatment of adrenal disorders can prevent or minimize growth disturbance and facilitate improved height gain. Mechanisms of growth disturbance in the following abnormal states will be discussed; hypercortisolaemia, hyperandrogenaemia and obesity. Prevalence and features of growth disturbance will be discussed in ACTH-dependent and ACTH-independent Cushing's syndrome, adrenocortical tumours, premature adrenarche, congenital adrenal hyperplasia and adrenal insufficiency disorders. Recommendations for management have been included.
Collapse
Affiliation(s)
- Marianna Minnetti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Silvana Caiulo
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
| | - Rosario Ferrigno
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | - Barbara Baldini-Ferroli
- Dipartimento Pediatrico Universitario Ospedaliero, Bambino Gesu' Children's Hospital, Rome, Italy
| | - Giorgia Bottaro
- Dipartimento Pediatrico Universitario Ospedaliero, Bambino Gesu' Children's Hospital, Rome, Italy
| | - Daniele Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Emilia Sbardella
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Martin O Savage
- Endocrinology Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, UK
| |
Collapse
|
37
|
Sayedyahossein S, Hedman AC, Sacks DB. Insulin suppresses transcriptional activity of yes-associated protein in insulin target cells. Mol Biol Cell 2020; 31:131-141. [PMID: 31693448 PMCID: PMC6960410 DOI: 10.1091/mbc.e19-04-0205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Yes-associated protein (YAP), the main transcriptional coactivator of the Hippo pathway, integrates multiple inputs from different signaling cascades. Evidence implicates YAP in the control of cellular nutrient and energy status, but the underlying mechanisms are not fully elucidated. Here we show that insulin modulates YAP transcriptional activity in classic insulin target cells, namely HepG2 and C2C12. Insulin increases YAP phosphorylation and significantly decreases YAP abundance in HepG2 cell nuclei. Proximity ligation assay analysis revealed a marked reduction in the interaction of YAP with TEA domain (TEAD) transcription factors in the nuclei of insulin-exposed cells. Consistent with these findings, insulin impaired both YAP/TEAD-mediated transcription and transcription of YAP target genes in HepG2 and C2C12 cells. Serum starvation abrogated the effect of insulin on YAP phosphorylation and YAP transcription. Both the expression of two gluconeogenesis genes, G6PC and PCK1, and the inhibitory effect of insulin on these genes were attenuated in YAP-deficient HepG2 cells. Our results identify insulin as a previously undescribed suppressor of YAP activity in insulin target cells and provide insight into cross-talk between the insulin and Hippo pathways.
Collapse
Affiliation(s)
- Samar Sayedyahossein
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, MD 20892
| | - Andrew C Hedman
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, MD 20892
| | - David B Sacks
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
38
|
Insulin Enhances the In Vitro Osteogenic Capacity of Flexor Tendon-Derived Progenitor Cells. Stem Cells Int 2019; 2019:1602751. [PMID: 31949435 PMCID: PMC6948345 DOI: 10.1155/2019/1602751] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 11/17/2019] [Accepted: 11/30/2019] [Indexed: 12/13/2022] Open
Abstract
There is increased incidence of tendon disorders and decreased tendon healing capacity in people with diabetes mellitus (DM). Recent studies have also suggested pathological ossification in repair tendon of people with DM. Therefore, the objective of this study is to investigate the effects of insulin supplementation, an important pathophysiologic stimulus of DM, on tendon progenitor cell (TPC) proliferation and in vitro osteogenic capacity. Passage 3 TPCs were isolated from collagenase-digested, equine superficial digital flexor tendons. TPC proliferation was measured via MTT assay after 3 days of monolayer culture in medium supplemented with 0, 0.007, 0.07, and 0.7 nM insulin. In vitro osteogenic capacity of TPCs (Alizarin Red staining, osteogenic mRNA expression, and alkaline phosphatase bioactivity) was assessed with 0, 0.07, and 0.7 nM insulin-supplemented osteogenic induction medium. Insulin (0.7 nM) significantly increased TPC proliferation after 3 days of monolayer culture. Alizarin Red staining of insulin-treated TPC osteogenic cultures demonstrated robust cell aggregation and mineralized matrix secretion, in a dose-dependent manner. Runx2, alkaline phosphatase, and Osteonectin mRNA and alkaline phosphatase bioactivity of insulin-treated TPC cultures were significantly higher at day 14 of osteogenesis compared to untreated controls. Addition of picropodophyllin (PPP), a selective IGF-I receptor inhibitor, mitigated the increased osteogenic capacity of TPCs, indicating that IGF-I signaling plays an important role. Our findings indicate that hyperinsulinemia may alter TPC phenotype and subsequently impact the quality of repair tendon tissue.
Collapse
|
39
|
Holly JMP, Biernacka K, Perks CM. The Neglected Insulin: IGF-II, a Metabolic Regulator with Implications for Diabetes, Obesity, and Cancer. Cells 2019; 8:cells8101207. [PMID: 31590432 PMCID: PMC6829378 DOI: 10.3390/cells8101207] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023] Open
Abstract
When originally discovered, one of the initial observations was that, when all of the insulin peptide was depleted from serum, the vast majority of the insulin activity remained and this was due to a single additional peptide, IGF-II. The IGF-II gene is adjacent to the insulin gene, which is a result of gene duplication, but has evolved to be considerably more complicated. It was one of the first genes recognised to be imprinted and expressed in a parent-of-origin specific manner. The gene codes for IGF-II mRNA, but, in addition, also codes for antisense RNA, long non-coding RNA, and several micro RNA. Recent evidence suggests that each of these have important independent roles in metabolic regulation. It has also become clear that an alternatively spliced form of the insulin receptor may be the principle IGF-II receptor. These recent discoveries have important implications for metabolic disorders and also for cancer, for which there is renewed acknowledgement of the importance of metabolic reprogramming.
Collapse
Affiliation(s)
- Jeff M P Holly
- Department of Translational Health Science, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Learning & Research Building, Southmead Hospital, Bristol, BS10 5NB, UK.
| | - Kalina Biernacka
- Department of Translational Health Science, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Learning & Research Building, Southmead Hospital, Bristol, BS10 5NB, UK
| | - Claire M Perks
- Department of Translational Health Science, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Learning & Research Building, Southmead Hospital, Bristol, BS10 5NB, UK
| |
Collapse
|
40
|
Liu S, Borgland SL. Insulin actions in the mesolimbic dopamine system. Exp Neurol 2019; 320:113006. [DOI: 10.1016/j.expneurol.2019.113006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/21/2019] [Accepted: 07/03/2019] [Indexed: 01/22/2023]
|
41
|
Hendley MA, Murphy KP, Isely C, Struckman HL, Annamalai P, Gower RM. The host response to poly(lactide-co-glycolide) scaffolds protects mice from diet induced obesity and glucose intolerance. Biomaterials 2019; 217:119281. [PMID: 31260882 DOI: 10.1016/j.biomaterials.2019.119281] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 06/12/2019] [Accepted: 06/12/2019] [Indexed: 12/21/2022]
Abstract
Underlying metabolic disease is poor adipose tissue function characterized by impaired glucose tolerance and low expression of health promoting adipokines. Currently, no treatments specifically target the adipose tissue and we are investigating polymer scaffolds for localized drug delivery as a therapeutic platform. In this work we implanted porous poly(lactide-co-glycolide) scaffolds into the epididymal fat of mice. Surprisingly, "empty" scaffolds decreased blood glucose levels in healthy mice as well as epididymal fat pad size. By injecting a fluorescent glucose tracer into mice, we determined that glucose uptake increases by 60% in epididymal fat pads with scaffolds; in contrast, glucose uptake was not elevated in other major metabolic organs, suggesting the enhanced glucose uptake at the scaffold implant site was responsible for decreased blood glucose levels. Histology indicated increased cellularity and tissue remodeling around the scaffold and we found increased expression of glucose transporter 1 and insulin-like growth factor 1, which are proteins involved in wound healing that can also modulate blood glucose levels through their promotion of glucose uptake. Regarding clinical translation, "empty" scaffolds decreased obesity and improved glucose tolerance in mice fed a high fat diet. These findings demonstrate increased cellular activity in the adipose tissue, such as that associated with the host response to biomaterial implant, is beneficial in mice suffering from metabolic complications of over nutrition, possibly because it mitigates the positive energy balance that leads to the obese, diabetic state. More broadly, this work reaffirms that in addition to the local host response typically investigated, biomaterial implant has systemic physiological effects and suggests that there may be implications for therapy.
Collapse
Affiliation(s)
- Michael A Hendley
- Biomedical Engineering Program, University of South Carolina, Columbia, SC, 29208, USA
| | - Kendall P Murphy
- Department of Chemical Engineering, University of South Carolina, Columbia, SC, 29208, USA
| | - Christopher Isely
- Department of Chemical Engineering, University of South Carolina, Columbia, SC, 29208, USA
| | - Heather L Struckman
- Biomedical Engineering Program, University of South Carolina, Columbia, SC, 29208, USA
| | - Prakasam Annamalai
- Department of Chemical Engineering, University of South Carolina, Columbia, SC, 29208, USA
| | - R Michael Gower
- Department of Chemical Engineering, University of South Carolina, Columbia, SC, 29208, USA; Biomedical Engineering Program, University of South Carolina, Columbia, SC, 29208, USA.
| |
Collapse
|
42
|
Lee KL, Aitken JF, Hsu HL, Williams GM, Brimble MA, Cooper GJS. Glucoregulatory activity of vesiculin in insulin sensitive and resistant mice. Peptides 2019; 116:1-7. [PMID: 31018156 DOI: 10.1016/j.peptides.2019.04.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/11/2019] [Accepted: 04/15/2019] [Indexed: 11/28/2022]
Abstract
Pancreatic islet-derived peptide hormones play key roles in the maintenance of systemic energy homeostasis and glucose balance and defects in their regulation are strongly implicated in the pathogenesis of obesity and diabetes. Peptides have also been used as lead compounds for therapeutics targeting metabolic disease. It is therefore important to understand the activity and function of islet hormones in both their target tissues and the whole organism. Insulin-like growth factor II (IGF-II) is an insulin homolog secreted by the islet β-cells. Vesiculin is a newly discovered peptide hormone, processed from IGF-II and secreted from islet β-cells in response to glucose. We postulated that vesiculin might act to regulate systemic glucose metabolism. Here we report our original investigations of vesiculin's activity in relation to glucoregulation. Vesiculin and IGF-II displayed similar dose-response relationships for lowering blood glucose in insulin-responsive FVB/n mice. By contrast, the ability of IGF-II to lower blood glucose was blunted in insulin-resistant triprolyl human-amylin transgenic mice, whereas vesiculin's ability to lower blood glucose remained unaffected. We also confirmed the ability of vesiculin to bypass insulin resistance in a second mouse model. In vitro analysis of signalling by vesiculin and IGF-II indicates that, like IGF-II, vesiculin signals through the IR/ IGF1R. Overall, we show that removal of only four amino acids from IGF-II has generated a peptide hormone with different bioactivity relevant to blood-glucose regulation. Investigating the differences among vesiculin, IGF-II and insulin signalling and activity may provide new insights into insulin resistance and potentially inform the design of novel therapeutics.
Collapse
Affiliation(s)
- Kate L Lee
- Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.; Maurice Wilkins Centre for Molecular BioDiscovery, Auckland, New Zealand..
| | - Jacqueline F Aitken
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Huai-L Hsu
- Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Geoffrey M Williams
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
| | - Margaret A Brimble
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand.; Maurice Wilkins Centre for Molecular BioDiscovery, Auckland, New Zealand
| | - Garth J S Cooper
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand.; Maurice Wilkins Centre for Molecular BioDiscovery, Auckland, New Zealand.; Centre for Advanced Discovery and Experimental Therapeutics, Manchester Biomedical Research Centre, Central Manchester University Hospitals NHS Foundation Trust, and the School of Biomedicine, the Medical School, University of Manchester, Manchester, UK
| |
Collapse
|
43
|
Paracrine Mechanisms of Redox Signalling for Postmitotic Cell and Tissue Regeneration. Trends Cell Biol 2019; 29:514-530. [DOI: 10.1016/j.tcb.2019.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/10/2019] [Accepted: 01/18/2019] [Indexed: 01/08/2023]
|
44
|
Zhu Y, Li X, Liu J, Zhou G, Yu Y, Jing L, Shi Z, Zhou X, Sun Z. The effects of decabromodiphenyl ether on glycolipid metabolism and related signaling pathways in mice. CHEMOSPHERE 2019; 222:849-855. [PMID: 30743236 DOI: 10.1016/j.chemosphere.2019.02.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 01/29/2019] [Accepted: 02/01/2019] [Indexed: 06/09/2023]
Abstract
Decabromodiphenyl ether (BDE-209), an addictive type flame retardant, is widely found in environments, and could affect the glycolipid metabolism. The present study was designed to investigate the potential mechanism of BDE-209 affecting glycolipid metabolism. Forty mice were randomly divided into four groups, and they were exposed to BDE-209 at dosages of 0, 7.5, 25 and 75 mg kg-1·d-1 for 28 d, respectively. The results showed that BDE-209 increased the serum levels of glucose, insulin, and triglyceride, also decreased the level of high-density lipoprotein, and damaged the structures of liver and adipose tissue in mice. BDE-209 significantly increased the protein expression of p-IRS, markedly decreased the expressions of PI3K, p-AKT, and GLUT4, significantly improved the lipid metabolism related factor expressions of p-mTOR, mTOR, PPARγ and RXRɑ, also inhibited the activity of antioxidant enzymes in the liver of mice. These results suggested that BDE-209 could affect glucose metabolism and inhibiting PI3K/AKT/GLUT4 signaling pathway resulting from improving the p-IRS expression, and interfered with lipid metabolism through activate mTOR/PPARγ/RXRα resulting from oxidative stress in mice.
Collapse
Affiliation(s)
- Yupeng Zhu
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, 100069, Beijing, China
| | - Xiangyang Li
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, 100069, Beijing, China
| | - Jianhui Liu
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, 100069, Beijing, China
| | - Guiqing Zhou
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, 100069, Beijing, China
| | - Yang Yu
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, 100069, Beijing, China.
| | - Li Jing
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, 100069, Beijing, China
| | - Zhixiong Shi
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, 100069, Beijing, China
| | - Xianqing Zhou
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, 100069, Beijing, China.
| | - Zhiwei Sun
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, 100069, Beijing, China
| |
Collapse
|
45
|
Solarek W, Koper M, Lewicki S, Szczylik C, Czarnecka AM. Insulin and insulin-like growth factors act as renal cell cancer intratumoral regulators. J Cell Commun Signal 2019; 13:381-394. [PMID: 30929166 PMCID: PMC6732138 DOI: 10.1007/s12079-019-00512-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/25/2019] [Indexed: 12/24/2022] Open
Abstract
The risk of renal cell carcinoma development is correlated with obesity and type II diabetes. Since insulin and insulin-like growth factors play a key role during development of both metabolic diseases, these molecules may be important in RCC pathophysiology We investigated the effect of insulin and IGFs on RCC cells using in vitro model with 786-O, 769-P, Caki-1, Caki-2, ACHN cancer cell lines. Cancer cells were compared with normal kidney cells - PCS-400-010 and HEK293. The growth, viability of cells as well as migration rate were assessed upon hormonal stimulation. The insulin receptor and Insulin-like growth factor 1 receptor presence were evaluated and the expression of 84 genes related to insulin signaling pathway. In all RCC cell lines IGF-1R expression was confirmed in contrast to IR, which was expressed only in control HEK293 cell line. Insulin and IGFs stimulated RCC cells growth and migration rate. Insulin, IGF-1 and IGF-2 triggered both IR and IGF-1R phosphorylation. Analyzed RCC did not secret insulin, IGF-1 or IGF-2 and were not activated in autocrine-paracrine signaling loop. Insulin and IGFs stimulations triggered down-regulation of PI3K-Akt-mTOR and Ras-MAPK pathway gens, as well as DOK2-3, INS, FRS3, IRS1-2, IGF1R - genes encoding insulin receptor-associated proteins. In conclusion, we showed that IGFs and insulin may play a stimulatory role for renal cancer cells, thus they can possibly affect renal cancer tumorigenesis and progression on cellular level.
Collapse
Affiliation(s)
- Wojciech Solarek
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, Warsaw, 04-141, Poland.,School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Michal Koper
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Slawomir Lewicki
- Department of Regenerative Medicine and Cell Biology, Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | - Cezary Szczylik
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, Warsaw, 04-141, Poland.,School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland.,Department of Oncology, European Health Centre, Otwock, Poland.,Medical Center for Postgraduate Education, Warsaw, Poland
| | - Anna M Czarnecka
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, Warsaw, 04-141, Poland.
| |
Collapse
|
46
|
Holly JMP, Biernacka K, Perks CM. Systemic Metabolism, Its Regulators, and Cancer: Past Mistakes and Future Potential. Front Endocrinol (Lausanne) 2019; 10:65. [PMID: 30809194 PMCID: PMC6380210 DOI: 10.3389/fendo.2019.00065] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/23/2019] [Indexed: 12/28/2022] Open
Abstract
There has been a resurgence of interest in cancer metabolism; primarily in the resetting of metabolism within malignant cells. Metabolism within cells has always been a tightly regulated process; initially in protozoans due to metabolic enzymes, and the intracellular signaling pathways that regulate these, being directly sensitive to the availability of nutrients. With the evolution of metazoans many of these controls had been overlaid by extra-cellular regulators that ensured coordinated regulation of metabolism within the community of cells that comprised the organism. Central to these systemic regulators is the insulin/insulin-like growth factor (IGF) system that throughout evolution has integrated the control of tissue growth with metabolic status. Oncological interest in the main systemic metabolic regulators greatly subsided when pharmaceutical strategies designed to treat cancers failed in the clinic. During the same period, however the explosion of new information from genetics has revealed the complexity and heterogeneity of advanced cancers and helped explain the problems of managing cancer when it reaches such a stage. Evidence has also accumulated implying that the setting of the internal environment determines whether cancers progress to advanced disease and metabolic status is clearly an important component of this local ecology. We are in the midst of an epidemic of metabolic disorders and there is considerable research into strategies for controlling metabolism. Integrating these new streams of information suggests new possibilities for cancer prevention; both primary and secondary.
Collapse
Affiliation(s)
- Jeff M. P. Holly
- Faculty of Medicine, School of Translational Health Science, University of Bristol, Southmead Hospital, Bristol, United Kingdom
| | | | | |
Collapse
|
47
|
Strange K, Yamada T, Denton JS. A 30-year journey from volume-regulated anion currents to molecular structure of the LRRC8 channel. J Gen Physiol 2019; 151:100-117. [PMID: 30651298 PMCID: PMC6363415 DOI: 10.1085/jgp.201812138] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/03/2019] [Indexed: 12/18/2022] Open
Abstract
Strange et al. review recent advances in our understanding of the molecular and structural basis of volume-regulated anion channel function within the framework of classical biophysical and physiological studies. The swelling-activated anion channel VRAC has fascinated and frustrated physiologists since it was first described in 1988. Multiple laboratories have defined VRAC’s biophysical properties and have shown that it plays a central role in cell volume regulation and possibly other fundamental physiological processes. However, confusion and intense controversy surrounding the channel’s molecular identity greatly hindered progress in the field for >15 yr. A major breakthrough came in 2014 with the demonstration that VRAC is a heteromeric channel encoded by five members of the Lrrc8 gene family, Lrrc8A–E. A mere 4 yr later, four laboratories described cryo-EM structures of LRRC8A homomeric channels. As the melee of structure/function and physiology studies begins, it is critical that this work be framed by a clear understanding of VRAC biophysics, regulation, and cellular physiology as well as by the field’s past confusion and controversies. That understanding is essential for the design and interpretation of structure/function studies, studies of VRAC physiology, and studies aimed at addressing the vexing problem of how the channel detects cell volume changes. In this review we discuss key aspects of VRAC biophysics, regulation, and function and integrate these into our emerging understanding of LRRC8 protein structure/function.
Collapse
Affiliation(s)
- Kevin Strange
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN.,Novo Biosciences, Inc., Bar Harbor, ME
| | - Toshiki Yamada
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN
| | - Jerod S Denton
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
48
|
Abstract
We present a protocol for building, validating, and simulating models of signal transduction networks. These networks are challenging modeling targets due to the combinatorial complexity and sparse data, which have made it a major challenge even to formalize the current knowledge. To address this, the community has developed methods to model biomolecular reaction networks based on site dynamics. The strength of this approach is that reactions and states can be defined at variable resolution, which makes it possible to adapt the model resolution to the empirical data. This improves both scalability and accuracy, making it possible to formalize large models of signal transduction networks. Here, we present a method to build and validate large models of signal transduction networks. The workflow is based on rxncon, the reaction-contingency language. In a five-step process, we create a mechanistic network model, convert it into an executable Boolean model, use the Boolean model to evaluate and improve the network, and finally export the rxncon model into a rule-based format. We provide an introduction to the rxncon language and an annotated, step-by-step protocol for the workflow. Finally, we create a small model of the insulin signaling pathway to illustrate the protocol, together with some of the challenges-and some of their solutions-in modeling signal transduction.
Collapse
Affiliation(s)
- Jesper Romers
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sebastian Thieme
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ulrike Münzner
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany.,Bioinformatics Center, Institute for Chemical Research, Kyoto University, Uji, Japan
| | - Marcus Krantz
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
49
|
Vassilakos G, Barton ER. Insulin-Like Growth Factor I Regulation and Its Actions in Skeletal Muscle. Compr Physiol 2018; 9:413-438. [PMID: 30549022 DOI: 10.1002/cphy.c180010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The insulin-like growth factor (IGF) pathway is essential for promoting growth and survival of virtually all tissues. It bears high homology to its related protein insulin, and as such, there is an interplay between these molecules with regard to their anabolic and metabolic functions. Skeletal muscle produces a significant proportion of IGF-1, and is highly responsive to its actions, including increased muscle mass and improved regenerative capacity. In this overview, the regulation of IGF-1 production, stability, and activity in skeletal muscle will be described. Second, the physiological significance of the forms of IGF-1 produced will be discussed. Last, the interaction of IGF-1 with other pathways will be addressed. © 2019 American Physiological Society. Compr Physiol 9:413-438, 2019.
Collapse
Affiliation(s)
- Georgios Vassilakos
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, USA
| | - Elisabeth R Barton
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
50
|
Zhou T, Ma Y, Tang J, Guo F, Dong M, Wei Q. Modulation of IGF1R Signaling Pathway by GIGYF1 in High Glucose-Induced SHSY-5Y Cells. DNA Cell Biol 2018; 37:1044-1054. [PMID: 30376373 DOI: 10.1089/dna.2018.4336] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Ting Zhou
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuefei Ma
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Juan Tang
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fengqi Guo
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mingxia Dong
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qianping Wei
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|