101
|
Kharade SV, Sanchez-Andres JV, Fulton MG, Shelton EL, Blobaum AL, Engers DW, Hofmann CS, Dadi PK, Lantier L, Jacobson DA, Lindsley CW, Denton JS. Structure-Activity Relationships, Pharmacokinetics, and Pharmacodynamics of the Kir6.2/SUR1-Specific Channel Opener VU0071063. J Pharmacol Exp Ther 2019; 370:350-359. [PMID: 31201216 PMCID: PMC6691189 DOI: 10.1124/jpet.119.257204] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 06/12/2019] [Indexed: 01/14/2023] Open
Abstract
Glucose-stimulated insulin secretion from pancreatic β-cells is controlled by ATP-regulated potassium (KATP) channels composed of Kir6.2 and sulfonylurea receptor 1 (SUR1) subunits. The KATP channel-opener diazoxide is FDA-approved for treating hyperinsulinism and hypoglycemia but suffers from off-target effects on vascular KATP channels and other ion channels. The development of more specific openers would provide critically needed tool compounds for probing the therapeutic potential of Kir6.2/SUR1 activation. Here, we characterize a novel scaffold activator of Kir6.2/SUR1 that our group recently discovered in a high-throughput screen. Optimization efforts with medicinal chemistry identified key structural elements that are essential for VU0071063-dependent opening of Kir6.2/SUR1. VU0071063 has no effects on heterologously expressed Kir6.1/SUR2B channels or ductus arteriole tone, indicating it does not open vascular KATP channels. VU0071063 induces hyperpolarization of β-cell membrane potential and inhibits insulin secretion more potently than diazoxide. VU0071063 exhibits metabolic and pharmacokinetic properties that are favorable for an in vivo probe and is brain penetrant. Administration of VU0071063 inhibits glucose-stimulated insulin secretion and glucose-lowering in mice. Taken together, these studies indicate that VU0071063 is a more potent and specific opener of Kir6.2/SUR1 than diazoxide and should be useful as an in vitro and in vivo tool compound for investigating the therapeutic potential of Kir6.2/SUR1 expressed in the pancreas and brain.
Collapse
Affiliation(s)
- Sujay V Kharade
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Juan Vicente Sanchez-Andres
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Mark G Fulton
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Elaine L Shelton
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Anna L Blobaum
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Darren W Engers
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Christopher S Hofmann
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Prasanna K Dadi
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Louise Lantier
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - David A Jacobson
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Craig W Lindsley
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Jerod S Denton
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| |
Collapse
|
102
|
Microbiota signatures relating to reduced memory and exploratory behaviour in the offspring of overweight mothers in a murine model. Sci Rep 2019; 9:12609. [PMID: 31471539 PMCID: PMC6717200 DOI: 10.1038/s41598-019-48090-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 07/18/2019] [Indexed: 12/26/2022] Open
Abstract
An elevated number of women of reproductive age are overweight, predisposing their offspring to metabolic and neuropsychiatric disorders. Gut microbiota is influenced by maternal factors, and has been implicated in the pathogenesis of neurodegenerative diseases. Our aim was to explore the effects of maternal high-fat feeding on the relationship linking gut microbiota and cognitive development in the offspring. Murine offspring born to dams undergoing normal diet (NDm) and high-fat diet (HFDm) were studied at 1 or 6 months of age to assess cognitive function by Y-maze test, cerebral glucose metabolism and insulin sensitivity by Positron Emission Tomography, brain density by Computed Tomography, microbiota profile (colon, caecum) and inferred metabolic pathways (KEGG analysis) by 16S ribosomal RNA sequencing. From 3 weeks post-weaning, mice born to HFDm developed hyperphagia and overweight, showing reduction in memory and exploratory behaviour, and brain insulin resistance in adulthood. We identified a panel of bacteria characterizing offspring born to HFD dams from early life, and correlating with dysfunction in memory and exploratory behaviour in adults (including Proteobacteria phylum, Parabacteroides and unclassified Rikenellaceae genera). Microbiota-derived metabolic pathways involved in fatty acid, essential aminoacid and vitamin processing, sulphur metabolism, glutaminergic activation and Alzheimer’s disease were differently present in the HFDm and NDm offspring groups. Our results document tight relationships between gut dysbiosis and memory and behavioural impairment in relation to maternal HFD. Persistent bacterial signatures induced by maternal HFD during infancy can influence cognition during adulthood, opening the possibility of microbiota-targeted strategies to contrast cognitive decline.
Collapse
|
103
|
Abstract
PURPOSE OF REVIEW Increased glucose production associated with hepatic insulin resistance contributes to the development of hyperglycemia in T2D. The molecular mechanisms accounting for increased glucose production remain controversial. Our aims were to review recent literature concerning molecular mechanisms regulating glucose production and to discuss these mechanisms in the context of physiological experiments and observations in humans and large animal models. RECENT FINDINGS Genetic intervention studies in rodents demonstrate that insulin can control hepatic glucose production through both direct effects on the liver, and through indirect effects to inhibit adipose tissue lipolysis and limit gluconeogenic substrate delivery. However, recent experiments in canine models indicate that the direct effects of insulin on the liver are dominant over the indirect effects to regulate glucose production. Recent molecular studies have also identified insulin-independent mechanisms by which hepatocytes sense intrahepatic carbohydrate levels to regulate carbohydrate disposal. Dysregulation of hepatic carbohydrate sensing systems may participate in increased glucose production in the development of diabetes.
Collapse
Affiliation(s)
- Ashot Sargsyan
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Mark A Herman
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA.
- Division of Diabetes, Endocrinology, and Metabolism, Duke University, Durham, NC, USA.
| |
Collapse
|
104
|
Abstract
The blood-brain barrier (BBB) was first noted for its ability to prevent the unregulated exchange of substances between the blood and the central nervous system (CNS). Over time, its characterization as an interface that enables regulated exchanges between the CNS and substances that are carried in the blood in a hormone-like fashion have emerged. Therefore, communication between the CNS, BBB and peripheral tissues has many endocrine-like properties. In this Review, I examine the various ways in which the BBB exhibits endocrine-related properties. The BBB is a target for hormones, such as leptin and insulin, that affect many of its functions. The BBB is also a secretory body, releasing substances either into the blood or the interstitial fluid of the brain. The BBB selectively allows classical and non-classical hormones entry to and exit from the CNS, thus allowing the CNS to be both an endocrine target and a secretory tissue. The BBB is affected by endocrine diseases such as diabetes mellitus and can cause or participate in endocrine diseases, including those related to thyroid hormones and obesity. The endocrine-like mechanisms of the BBB can extend the definition of endocrine disease to include neurodegenerative conditions, including Alzheimer disease, and of hormones to include cytokines, triglycerides and fatty acids.
Collapse
Affiliation(s)
- William A Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
105
|
Kelly L, Almutairi MM, Kursan S, Pacheco R, Dias-Junior E, Castrop H, Di Fulvio M. Impaired glucose tolerance, glucagon, and insulin responses in mice lacking the loop diuretic-sensitive Nkcc2a transporter. Am J Physiol Cell Physiol 2019; 317:C843-C856. [PMID: 31365295 DOI: 10.1152/ajpcell.00144.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The Na+K+2Cl- cotransporter-2 (Nkcc2, Slc12a1) is abundantly expressed in the kidney and its inhibition with the loop-diuretics bumetanide and furosemide has been linked to transient or permanent hyperglycemia in mice and humans. Notably, Slc12a1 is expressed at low levels in hypothalamic neurons and in insulin-secreting β-cells of the endocrine pancreas. The present study was designed to determine if global elimination of one of the Slc12a1 products, i.e., Nkcc2 variant a (Nkcc2a), the main splice version of Nkcc2 found in insulin-secreting β-cells, has an impact on the insulin and glucagon secretory responses and fuel homeostasis in vivo. We have used dynamic tests of glucose homeostasis in wild-type mice and mice lacking both alleles of Nkcc2a (Nkcc2aKO) and assessed their islet secretory responses in vitro. Under basal conditions, Nkcc2aKO mice have impaired glucose homeostasis characterized by increased blood glucose, intolerance to the sugar, delayed/blunted in vivo insulin and glucagon responses to glucose, and increased glycemic responses to the gluconeogenic substrate alanine. Further, we provide evidence of conserved quantitative secretory responses of Nkcc2aKO islets within a context of increased islet size related to hyperplastic/hypertrophic glucagon- and insulin-positive cells (α-cells and β-cells, respectively), normal total islet Cl- content, and reduced β-cell expression of the Cl- extruder Kcc2.
Collapse
Affiliation(s)
- Lisa Kelly
- Department of Pharmacology and Toxicology, Wright State University, Dayton, Ohio
| | - Mohammed M Almutairi
- Department of Pharmacology and Toxicology, Wright State University, Dayton, Ohio
| | - Shams Kursan
- Department of Pharmacology and Toxicology, Wright State University, Dayton, Ohio
| | - Romario Pacheco
- Department of Pharmacology and Toxicology, Wright State University, Dayton, Ohio
| | - Eduardo Dias-Junior
- Department of Pharmacology and Toxicology, Wright State University, Dayton, Ohio
| | - Hayo Castrop
- Institute of Physiology, University of Regensburg, Regensburg Germany
| | - Mauricio Di Fulvio
- Department of Pharmacology and Toxicology, Wright State University, Dayton, Ohio
| |
Collapse
|
106
|
Qiu J, Bosch MA, Zhang C, Rønnekleiv OK, Kelly MJ. Estradiol Protects Neuropeptide Y/Agouti-Related Peptide Neurons against Insulin Resistance in Females. Neuroendocrinology 2019; 110:105-118. [PMID: 31212279 PMCID: PMC6920578 DOI: 10.1159/000501560] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 06/17/2019] [Indexed: 12/18/2022]
Abstract
When it comes to obesity, men exhibit a higher incidence of metabolic syndrome than women in early adult life, but this sex advantage wanes in postmenopausal women. A key diagnostic of the metabolic syndrome is insulin resistance in both peripheral tissues and brain, especially in the hypothalamus. Since the anorexigenic hormone 17β-estradiol (E2) regulates food intake in part by inhibiting the excitability of the hypothalamic neuropeptide Y/agouti-related peptide (NPY/AgRP) neurons, we hypothesized that E2 would protect against insulin resistance in NPY/AgRP neurons with diet-induced obesity (DIO). Therefore, we did whole-cell recordings and single cell quantitative polymerase chain reaction in arcuate NPYGFP neurons from both female and male mice to test the efficacy of insulin with DIO. The resting membrane potential and input resistance of NPY/AgRP neurons were significantly increased in DIO versus control-diet fed males. Most notably, the efficacy of insulin to activate KATP channels in NPY/AgRP neurons was significantly attenuated, although the KATP channel opener diazoxide was fully effective in NPY/AgRP neurons from DIO males, indicating that the KATP channels were expressed and functional. In contrast, insulin was fully efficacious to activate KATP channels in DIO females, and the response was reversed by the KATP channel blocker tolbutamide. However, the ability of insulin to activate KATP channels was abrogated with ovariectomy but fully restored with E2 replacement. Insulin resistance in obese males was likely mediated by an increase in suppressor of cytokine signaling-3 (SOCS-3), protein tyrosine phosphatase B (PTP1B) and T-cell protein tyrosine phosphatase (TCPTP) activity, since the expression of all 3 mRNAs were upregulated in the obese males but not in females. As proof of principle, pre-incubation of hypothalamic slices from DIO males with the PTP1B/TCPTP inhibitor CX08005 completely rescued the effects of insulin. Therefore, E2 protects NPY/AgRP neurons in females against insulin resistance through, at least in part, attenuating phosphatase activity. The neuroprotective effects of E2 may explain sex differences in the expression of metabolic syndrome that disappears with the loss of E2 in aging.
Collapse
Affiliation(s)
- Jian Qiu
- Department of Physiology and Pharmacology, Oregon Health
& Science University, Portland, Oregon, USA
| | - Martha A. Bosch
- Department of Physiology and Pharmacology, Oregon Health
& Science University, Portland, Oregon, USA
| | - Chunguang Zhang
- Department of Physiology and Pharmacology, Oregon Health
& Science University, Portland, Oregon, USA
| | - Oline K. Rønnekleiv
- Department of Physiology and Pharmacology, Oregon Health
& Science University, Portland, Oregon, USA
- Division of Neuroscience, National Primate Research Center,
Oregon Health & Science University, Beaverton, Oregon, USA
| | - Martin J. Kelly
- Department of Physiology and Pharmacology, Oregon Health
& Science University, Portland, Oregon, USA
- Division of Neuroscience, National Primate Research Center,
Oregon Health & Science University, Beaverton, Oregon, USA
| |
Collapse
|
107
|
Ojha A, Ojha U, Mohammed R, Chandrashekar A, Ojha H. Current perspective on the role of insulin and glucagon in the pathogenesis and treatment of type 2 diabetes mellitus. Clin Pharmacol 2019; 11:57-65. [PMID: 31191043 PMCID: PMC6515536 DOI: 10.2147/cpaa.s202614] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/28/2019] [Indexed: 01/17/2023] Open
Abstract
According to the World Health Organization, 422 million adults worldwide live with diabetes mellitus (DM), a significant portion of whom have type 2 diabetes. The discovery of insulin as a key regulator of glucose metabolism has revolutionized our understanding of DM and provided several therapeutic avenues. Most studies have so far predominantly focused on the role of insulin in type 2 diabetes. However, the balance between insulin and glucagon is essential in ensuring glucose homeostasis. In this review, we begin by evaluating the principal differences between insulin and glucagon with regard to their mechanism and control of their secretion. Next, we discuss their mode of action and effects on metabolism. We further explore how the two hormones impact the natural history of type 2 diabetes. Finally, we outline how current and emerging pharmacological agents attempt to exploit the properties of insulin and glucagon to benefit patients with type 2 diabetes.
Collapse
Affiliation(s)
- Ashutosh Ojha
- Shobhaben Pratapbhai Patel School Of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies, Mumbai, India
| | - Utkarsh Ojha
- Faculty of Medicine, Imperial College London, London, UK
| | - Raihan Mohammed
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Harsh Ojha
- Department of Life Sciences, University of Warwick, Coventry, UK
| |
Collapse
|
108
|
Agarwal SM, Caravaggio F, Costa-Dookhan KA, Castellani L, Kowalchuk C, Asgariroozbehani R, Graff-Guerrero A, Hahn M. Brain insulin action in schizophrenia: Something borrowed and something new. Neuropharmacology 2019; 163:107633. [PMID: 31077731 DOI: 10.1016/j.neuropharm.2019.05.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/15/2019] [Accepted: 05/07/2019] [Indexed: 12/24/2022]
Abstract
Insulin signaling in the central nervous system is at the intersection of brain and body interactions, and represents a fundamental link between metabolic and cognitive disorders. Abnormalities in brain insulin action could underlie the development of comorbid schizophrenia and type 2 diabetes. Among its functions, central nervous system insulin is involved in regulation of striatal dopamine levels, peripheral glucose homeostasis, and feeding regulation. In this review, we discuss the role and importance of central nervous system insulin in schizophrenia and diabetes pathogenesis from a historical and mechanistic perspective. We describe central nervous system insulin sites and pathways of action, with special emphasis on glucose metabolism, cognitive functioning, inflammation, and food preferences. Finally, we suggest possible mechanisms that may explain the actions of central nervous system insulin in relation to schizophrenia and diabetes, focusing on glutamate and dopamine signaling, intracellular signal transduction pathways, and brain energetics. Understanding the interplay between central nervous system insulin and schizophrenia is essential to disentangling this comorbid relationship and may provide novel treatment approaches for both neuropsychiatric and metabolic dysfunction. This article is part of the issue entitled 'Special Issue on Antipsychotics'.
Collapse
Affiliation(s)
- Sri Mahavir Agarwal
- Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Fernando Caravaggio
- Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Kenya A Costa-Dookhan
- Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Chantel Kowalchuk
- Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Ariel Graff-Guerrero
- Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Margaret Hahn
- Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
109
|
Cedernaes J, Huang W, Ramsey KM, Waldeck N, Cheng L, Marcheva B, Omura C, Kobayashi Y, Peek CB, Levine DC, Dhir R, Awatramani R, Bradfield CA, Wang XA, Takahashi JS, Mokadem M, Ahima RS, Bass J. Transcriptional Basis for Rhythmic Control of Hunger and Metabolism within the AgRP Neuron. Cell Metab 2019; 29:1078-1091.e5. [PMID: 30827863 PMCID: PMC6506361 DOI: 10.1016/j.cmet.2019.01.023] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 11/12/2018] [Accepted: 01/30/2019] [Indexed: 12/12/2022]
Abstract
The alignment of fasting and feeding with the sleep/wake cycle is coordinated by hypothalamic neurons, though the underlying molecular programs remain incompletely understood. Here, we demonstrate that the clock transcription pathway maximizes eating during wakefulness and glucose production during sleep through autonomous circadian regulation of NPY/AgRP neurons. Tandem profiling of whole-cell and ribosome-bound mRNAs in morning and evening under dynamic fasting and fed conditions identified temporal control of activity-dependent gene repertoires in AgRP neurons central to synaptogenesis, bioenergetics, and neurotransmitter and peptidergic signaling. Synaptic and circadian pathways were specific to whole-cell RNA analyses, while bioenergetic pathways were selectively enriched in the ribosome-bound transcriptome. Finally, we demonstrate that the AgRP clock mediates the transcriptional response to leptin. Our results reveal that time-of-day restriction in transcriptional control of energy-sensing neurons underlies the alignment of hunger and food acquisition with the sleep/wake state.
Collapse
Affiliation(s)
- Jonathan Cedernaes
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Medical Sciences, Uppsala University, Uppsala SE-75124, Sweden
| | - Wenyu Huang
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kathryn Moynihan Ramsey
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Nathan Waldeck
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Lei Cheng
- Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL 60208, USA
| | - Biliana Marcheva
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Chiaki Omura
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yumiko Kobayashi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Clara Bien Peek
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Daniel C Levine
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ravindra Dhir
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Raj Awatramani
- Department of Neurology and Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Christopher A Bradfield
- McArdle Laboratory for Cancer Research, University of Wisconsin Medical School, Madison, WI 53706, USA
| | - Xiaozhong A Wang
- Department of Molecular Sciences, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL 60208, USA
| | - Joseph S Takahashi
- Department of Neuroscience and Howard Hughes Medical Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mohamad Mokadem
- Division of Gastroenterology and Hepatology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa, IA 52242, USA
| | - Rexford S Ahima
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Joseph Bass
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
110
|
Lundqvist MH, Almby K, Abrahamsson N, Eriksson JW. Is the Brain a Key Player in Glucose Regulation and Development of Type 2 Diabetes? Front Physiol 2019; 10:457. [PMID: 31133864 PMCID: PMC6524713 DOI: 10.3389/fphys.2019.00457] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/01/2019] [Indexed: 01/08/2023] Open
Abstract
Ever since Claude Bernards discovery in the mid 19th-century that a lesion in the floor of the third ventricle in dogs led to altered systemic glucose levels, a role of the CNS in whole-body glucose regulation has been acknowledged. However, this finding was later overshadowed by the isolation of pancreatic hormones in the 20th century. Since then, the understanding of glucose homeostasis and pathology has primarily evolved around peripheral mechanism. Due to scientific advances over these last few decades, however, increasing attention has been given to the possibility of the brain as a key player in glucose regulation and the pathogenesis of metabolic disorders such as type 2 diabetes. Studies of animals have enabled detailed neuroanatomical mapping of CNS structures involved in glucose regulation and key neuronal circuits and intracellular pathways have been identified. Furthermore, the development of neuroimaging techniques has provided methods to measure changes of activity in specific CNS regions upon diverse metabolic challenges in humans. In this narrative review, we discuss the available evidence on the topic. We conclude that there is much evidence in favor of active CNS involvement in glucose homeostasis but the relative importance of central vs. peripheral mechanisms remains to be elucidated. An increased understanding of this field may lead to new CNS-focusing pharmacologic strategies in the treatment of type 2 diabetes.
Collapse
Affiliation(s)
| | - Kristina Almby
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Jan W Eriksson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
111
|
Chang EH, Chavan SS, Pavlov VA. Cholinergic Control of Inflammation, Metabolic Dysfunction, and Cognitive Impairment in Obesity-Associated Disorders: Mechanisms and Novel Therapeutic Opportunities. Front Neurosci 2019; 13:263. [PMID: 31024226 PMCID: PMC6460483 DOI: 10.3389/fnins.2019.00263] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/06/2019] [Indexed: 12/26/2022] Open
Abstract
Obesity and obesity-associated disorders have become world-wide epidemics, substantially increasing the risk of debilitating morbidity and mortality. A characteristic feature of these disorders, which include the metabolic syndrome (MetS) and type 2 diabetes, is chronic low-grade inflammation stemming from metabolic and immune dysregulation. Inflammation in the CNS (neuroinflammation) and cognitive impairment have also been associated with obesity-driven disorders. The nervous system has a documented role in the regulation of metabolic homeostasis and immune function, and recent studies have indicated the important role of vagus nerve and brain cholinergic signaling in this context. In this review, we outline relevant aspects of this regulation with a specific focus on obesity-associated conditions. We outline accumulating preclinical evidence for the therapeutic efficacy of cholinergic stimulation in alleviating obesity-associated inflammation, neuroinflammation, and metabolic derangements. Recently demonstrated beneficial effects of galantamine, a centrally acting cholinergic drug and cognitive enhancer, in patients with MetS are also summarized. These studies provide a rationale for further therapeutic developments using pharmacological and bioelectronic cholinergic modulation for clinical benefit in obesity-associated disorders.
Collapse
Affiliation(s)
- Eric H. Chang
- Center for Bioelectronic Medicine and Biomedical Sciences, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Sangeeta S. Chavan
- Center for Bioelectronic Medicine and Biomedical Sciences, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Valentin A. Pavlov
- Center for Bioelectronic Medicine and Biomedical Sciences, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| |
Collapse
|
112
|
Wardzinski EK, Friedrichsen L, Dannenberger S, Kistenmacher A, Melchert UH, Jauch-Chara K, Oltmanns KM. Double transcranial direct current stimulation of the brain increases cerebral energy levels and systemic glucose tolerance in men. J Neuroendocrinol 2019; 31:e12688. [PMID: 30659676 DOI: 10.1111/jne.12688] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 12/14/2018] [Accepted: 01/15/2019] [Indexed: 12/31/2022]
Abstract
Transcranial direct current stimulation (tDCS) is a neuromodulatory method that has been tested experimentally and has already been used as an adjuvant therapeutic option to treat a number of neurological disorders and neuropsychiatric diseases. Beyond its well known local effects within the brain, tDCS also transiently promotes systemic glucose uptake and reduces the activity of the neurohormonal stress axes. We aimed to test whether the effects of a single tDCS application could be replicated upon double stimulation to persistently improve systemic glucose tolerance and stress axes activity in humans. In a single-blinded cross-over study, we examined 15 healthy male volunteers. Anodal tDCS vs sham was applied twice in series. Systemic glucose tolerance was investigated by the standard hyperinsulinaemic-euglycaemic glucose clamp procedure, and parameters of neurohormonal stress axes activity were measured. Because tDCS-induced brain energy consumption has been shown to be part of the mechanism underlying the assumed effects, we monitored the cerebral high-energy phosphates ATP and phosphocreatine by 31 phosphorus magnetic resonance spectroscopy. As hypothesised, analyses revealed that double anodal tDCS persistently increases glucose tolerance compared to sham. Moreover, we observed a significant rise in cerebral high-energy phosphate content upon double tDCS. Accordingly, the activity of the neurohormonal stress axes was reduced upon tDCS compared to sham. Our data demonstrate that double tDCS promotes systemic glucose uptake and reduces stress axes activity in healthy humans. These effects suggest that repetitive tDCS may be a future non-pharmacological option for combating glucose intolerance in type 2 diabetes patients.
Collapse
Affiliation(s)
- Ewelina K Wardzinski
- Section of Psychoneurobiology, Center of Brain, Behavior and Metabolism, University of Luebeck, Luebeck, Germany
| | - Lisa Friedrichsen
- Section of Psychoneurobiology, Center of Brain, Behavior and Metabolism, University of Luebeck, Luebeck, Germany
| | - Sina Dannenberger
- Section of Psychoneurobiology, Center of Brain, Behavior and Metabolism, University of Luebeck, Luebeck, Germany
| | - Alina Kistenmacher
- Section of Psychoneurobiology, Center of Brain, Behavior and Metabolism, University of Luebeck, Luebeck, Germany
| | - Uwe H Melchert
- Section of Psychoneurobiology, Center of Brain, Behavior and Metabolism, University of Luebeck, Luebeck, Germany
| | - Kamila Jauch-Chara
- Section of Psychoneurobiology, Center of Brain, Behavior and Metabolism, University of Luebeck, Luebeck, Germany
| | - Kerstin M Oltmanns
- Section of Psychoneurobiology, Center of Brain, Behavior and Metabolism, University of Luebeck, Luebeck, Germany
| |
Collapse
|
113
|
Lyra E Silva NDM, Gonçalves RA, Boehnke SE, Forny-Germano L, Munoz DP, De Felice FG. Understanding the link between insulin resistance and Alzheimer's disease: Insights from animal models. Exp Neurol 2019; 316:1-11. [PMID: 30930096 DOI: 10.1016/j.expneurol.2019.03.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease affecting millions of people worldwide. AD is characterized by a profound impairment of higher cognitive functions and still lacks any effective disease-modifying treatment. Defective insulin signaling has been implicated in AD pathophysiology, but the mechanisms underlying this process are not fully understood. Here, we review the molecular mechanisms underlying defective brain insulin signaling in rodent models of AD, and in a non-human primate (NHP) model of the disease that recapitulates features observed in AD brains. We further highlight similarities between the NHP and human brains and discuss why NHP models of AD are important to understand disease mechanisms and to improve the translation of effective therapies to humans. We discuss how studies using different animal models have contributed to elucidate the link between insulin resistance and AD.
Collapse
Affiliation(s)
| | | | - Susan E Boehnke
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Leticia Forny-Germano
- Institute of Medical Biochemistry Leopoldo De Meis, Federal University of Rio de Janeiro, Brazil
| | - Douglas P Munoz
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada.
| | - Fernanda G De Felice
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada; Department of Psychiatry, Queen's University, Kingston, ON, Canada; Institute of Medical Biochemistry Leopoldo De Meis, Federal University of Rio de Janeiro, Brazil.
| |
Collapse
|
114
|
Santiago JCP, Hallschmid M. Outcomes and clinical implications of intranasal insulin administration to the central nervous system. Exp Neurol 2019; 317:180-190. [PMID: 30885653 DOI: 10.1016/j.expneurol.2019.03.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/12/2019] [Accepted: 03/13/2019] [Indexed: 12/20/2022]
Abstract
Insulin signaling in the brain plays a critical role in metabolic control and cognitive function. Targeting insulinergic pathways in the central nervous system via peripheral insulin administration is feasible, but associated with systemic effects that necessitate tight supervision or countermeasures. The intranasal route of insulin administration, which largely bypasses the circulation and thereby greatly reduces these obstacles, has now been repeatedly tested in proof-of-concept studies in humans as well as animals. It is routinely used in experimental settings to investigate the impact on eating behavior, peripheral metabolism, memory function and brain activation of acute or long-term enhancements in central nervous system insulin signaling. Epidemiological and experimental evidence linking deteriorations in metabolic control such as diabetes with neurodegenerative diseases imply pathophysiological relevance of dysfunctional brain insulin signaling or brain insulin resistance, and suggest that targeting insulin in the brain holds some promise as a therapy or adjunct therapy. This short narrative review gives an overview over recent findings on brain insulin signaling as derived from human studies deploying intranasal insulin, and evaluates the potential of therapeutic interventions that target brain insulin resistance.
Collapse
Affiliation(s)
- João C P Santiago
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, 72076 Tübingen, Germany; German Center for Diabetes Research (DZD), 72076 Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
| | - Manfred Hallschmid
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, 72076 Tübingen, Germany; German Center for Diabetes Research (DZD), 72076 Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany.
| |
Collapse
|
115
|
Edgerton DS, Scott M, Farmer B, Williams PE, Madsen P, Kjeldsen T, Brand CL, Fledelius C, Nishimura E, Cherrington AD. Targeting insulin to the liver corrects defects in glucose metabolism caused by peripheral insulin delivery. JCI Insight 2019; 5:126974. [PMID: 30830873 DOI: 10.1172/jci.insight.126974] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Peripheral hyperinsulinemia resulting from subcutaneous insulin injection is associated with metabolic defects which include abnormal glucose metabolism. The first aim of this study was to quantify the impairments in liver and muscle glucose metabolism that occur when insulin is delivered via a peripheral vein compared to when it is given through its endogenous secretory route (the hepatic portal vein) in overnight fasted conscious dogs. The second aim was to determine if peripheral delivery of a hepato-preferential insulin analog could restore the physiologic response to insulin that occurs under meal feeding conditions. This study is the first to show that hepatic glucose uptake correlates with insulin's direct effects on the liver under hyperinsulinemic-hyperglycemic conditions. In addition, glucose uptake was equally divided between the liver and muscle when insulin was infused into the portal vein, but when it was delivered into a peripheral vein the percentage of glucose taken up by muscle was 4-times greater than that going to the liver, with liver glucose uptake being less than half of normal. These defects could not be corrected by adjusting the dose of peripheral insulin. On the other hand, hepatic and non-hepatic glucose metabolism could be fully normalized by a hepato-preferential insulin analog.
Collapse
Affiliation(s)
- Dale S Edgerton
- Vanderbilt University School of Medicine, Department of Molecular Physiology and Biophysics, Nashville, Tennessee, USA
| | - Melanie Scott
- Vanderbilt University School of Medicine, Department of Molecular Physiology and Biophysics, Nashville, Tennessee, USA
| | - Ben Farmer
- Vanderbilt University School of Medicine, Department of Molecular Physiology and Biophysics, Nashville, Tennessee, USA
| | - Phillip E Williams
- Vanderbilt University Medical Center, Division of Surgical Research, Nashville, Tennessee, USA
| | - Peter Madsen
- Research and Development, Novo Nordisk A/S, Novo Nordisk Park, Maaleov, Denmark
| | - Thomas Kjeldsen
- Research and Development, Novo Nordisk A/S, Novo Nordisk Park, Maaleov, Denmark
| | - Christian L Brand
- Research and Development, Novo Nordisk A/S, Novo Nordisk Park, Maaleov, Denmark
| | - Christian Fledelius
- Research and Development, Novo Nordisk A/S, Novo Nordisk Park, Maaleov, Denmark
| | - Erica Nishimura
- Research and Development, Novo Nordisk A/S, Novo Nordisk Park, Maaleov, Denmark
| | - Alan D Cherrington
- Vanderbilt University School of Medicine, Department of Molecular Physiology and Biophysics, Nashville, Tennessee, USA
| |
Collapse
|
116
|
Desmoulins L, Chrétien C, Paccoud R, Collins S, Cruciani-Guglielmacci C, Galinier A, Liénard F, Quinault A, Grall S, Allard C, Fenech C, Carneiro L, Mouillot T, Fournel A, Knauf C, Magnan C, Fioramonti X, Pénicaud L, Leloup C. Mitochondrial Dynamin-Related Protein 1 (DRP1) translocation in response to cerebral glucose is impaired in a rat model of early alteration in hypothalamic glucose sensing. Mol Metab 2019; 20:166-177. [PMID: 30553770 PMCID: PMC6358535 DOI: 10.1016/j.molmet.2018.11.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 11/19/2018] [Accepted: 11/22/2018] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE Hypothalamic glucose sensing (HGS) initiates insulin secretion (IS) via a vagal control, participating in energy homeostasis. This requires mitochondrial reactive oxygen species (mROS) signaling, dependent on mitochondrial fission, as shown by invalidation of the hypothalamic DRP1 protein. Here, our objectives were to determine whether a model with a HGS defect induced by a short, high fat-high sucrose (HFHS) diet in rats affected the fission machinery and mROS signaling within the mediobasal hypothalamus (MBH). METHODS Rats fed a HFHS diet for 3 weeks were compared with animals fed a normal chow. Both in vitro (calcium imaging) and in vivo (vagal nerve activity recordings) experiments to measure the electrical activity of isolated MBH gluco-sensitive neurons in response to increased glucose level were performed. In parallel, insulin secretion to a direct glucose stimulus in isolated islets vs. insulin secretion resulting from brain glucose stimulation was evaluated. Intra-carotid glucose load-induced hypothalamic DRP1 translocation to mitochondria and mROS (H2O2) production were assessed in both groups. Finally, compound C was intracerebroventricularly injected to block the proposed AMPK-inhibited DRP1 translocation in the MBH to reverse the phenotype of HFHS fed animals. RESULTS Rats fed a HFHS diet displayed a decreased HGS-induced IS. Responses of MBH neurons to glucose exhibited an alteration of their electrical activity, whereas glucose-induced insulin secretion in isolated islets was not affected. These MBH defects correlated with a decreased ROS signaling and glucose-induced translocation of the fission protein DRP1, as the vagal activity was altered. AMPK-induced inhibition of DRP1 translocation increased in this model, but its reversal through the injection of the compound C, an AMPK inhibitor, failed to restore HGS-induced IS. CONCLUSIONS A hypothalamic alteration of DRP1-induced fission and mROS signaling in response to glucose was observed in HGS-induced IS of rats exposed to a 3 week HFHS diet. Early hypothalamic modifications of the neuronal activity could participate in a primary defect of the control of IS and ultimately, the development of diabetes.
Collapse
Affiliation(s)
- Lucie Desmoulins
- Centre des Sciences du Goût et de l'Alimentation, UMR CNRS 6265, INRA 1324, AgroSup, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France.
| | - Chloé Chrétien
- Centre des Sciences du Goût et de l'Alimentation, UMR CNRS 6265, INRA 1324, AgroSup, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France.
| | - Romain Paccoud
- Centre des Sciences du Goût et de l'Alimentation, UMR CNRS 6265, INRA 1324, AgroSup, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France.
| | - Stephan Collins
- Centre des Sciences du Goût et de l'Alimentation, UMR CNRS 6265, INRA 1324, AgroSup, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France.
| | - Céline Cruciani-Guglielmacci
- CNRS UMR 8251, Unit of Functional and Adaptive Biology, Paris, France; Department of Physiology, Université Paris Diderot, Paris, France.
| | - Anne Galinier
- STROMALab, UMR CNRS 5273, EFS Pyrénées-Méditerranée, Université Paul Sabatier, Toulouse, France.
| | - Fabienne Liénard
- Centre des Sciences du Goût et de l'Alimentation, UMR CNRS 6265, INRA 1324, AgroSup, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France.
| | - Aurore Quinault
- Centre des Sciences du Goût et de l'Alimentation, UMR CNRS 6265, INRA 1324, AgroSup, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France.
| | - Sylvie Grall
- Centre des Sciences du Goût et de l'Alimentation, UMR CNRS 6265, INRA 1324, AgroSup, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France.
| | - Camille Allard
- Centre des Sciences du Goût et de l'Alimentation, UMR CNRS 6265, INRA 1324, AgroSup, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France.
| | - Claire Fenech
- Centre des Sciences du Goût et de l'Alimentation, UMR CNRS 6265, INRA 1324, AgroSup, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France.
| | - Lionel Carneiro
- Centre des Sciences du Goût et de l'Alimentation, UMR CNRS 6265, INRA 1324, AgroSup, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France.
| | - Thomas Mouillot
- Centre des Sciences du Goût et de l'Alimentation, UMR CNRS 6265, INRA 1324, AgroSup, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France; Service d'Hépato-Gastroentérologie, hôpital du Bocage, Dijon, France.
| | - Audren Fournel
- Institut de Recherche en Santé Digestive, INSERM U1220, Université Paul Sabatier, Toulouse, France.
| | - Claude Knauf
- Institut de Recherche en Santé Digestive, INSERM U1220, Université Paul Sabatier, Toulouse, France.
| | - Christophe Magnan
- CNRS UMR 8251, Unit of Functional and Adaptive Biology, Paris, France.
| | - Xavier Fioramonti
- Centre des Sciences du Goût et de l'Alimentation, UMR CNRS 6265, INRA 1324, AgroSup, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France; UMR 1286, NutriNeuro, INRA, Université de Bordeaux, Bordeaux INP, Bordeaux, France.
| | - Luc Pénicaud
- Centre des Sciences du Goût et de l'Alimentation, UMR CNRS 6265, INRA 1324, AgroSup, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France.
| | - Corinne Leloup
- Centre des Sciences du Goût et de l'Alimentation, UMR CNRS 6265, INRA 1324, AgroSup, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France.
| |
Collapse
|
117
|
Loehfelm A, Boucsein A, Pretz D, Tups A. Timing Matters: Circadian Effects on Energy Homeostasis and Alzheimer's Disease. Trends Endocrinol Metab 2019; 30:132-143. [PMID: 30594436 DOI: 10.1016/j.tem.2018.12.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/09/2018] [Accepted: 12/03/2018] [Indexed: 10/27/2022]
Abstract
Metabolic syndrome and Alzheimer's disease (AD) are two major health issues in modern society causing an extraordinary financial burden for the global healthcare systems. A tight link between the pathologies of obesity and type 2 diabetes (T2D), and more recently between T2D and AD, has been discovered. Furthermore, in recent years it has become apparent that the circadian clock has an important function in controlling metabolism. This review integrates the role of the circadian clock in the development of these metabolic derangements and vice versa. Common features such as central insulin resistance, altered glycogen synthase kinase 3β (GSK3β) signalling, and central inflammation are discussed, and therapeutic interventions targeting those mechanisms are mentioned briefly.
Collapse
Affiliation(s)
- Aline Loehfelm
- Centre for Neuroendocrinology and Brain Health Research Centre, Department of Physiology, School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand; These authors contributed equally to this work
| | - Alisa Boucsein
- Centre for Neuroendocrinology and Brain Health Research Centre, Department of Physiology, School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand; These authors contributed equally to this work
| | - Dominik Pretz
- Centre for Neuroendocrinology and Brain Health Research Centre, Department of Physiology, School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand; These authors contributed equally to this work
| | - Alexander Tups
- Centre for Neuroendocrinology and Brain Health Research Centre, Department of Physiology, School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand.
| |
Collapse
|
118
|
Xu F, Yang J, Negishi H, Sun Y, Li D, Zhang X, Hayashi T, Gao M, Ikeda K, Ikejima T. Silibinin decreases hepatic glucose production through the activation of gut-brain-liver axis in diabetic rats. Food Funct 2019; 9:4926-4935. [PMID: 30178798 DOI: 10.1039/c8fo00565f] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Silibinin, a flavonolignan derived from milk thistle (Silybum marianum), has been revealed to have a beneficial effect on improving diabetes-impaired glycemic control. However, the underlying mechanism is still unclear. In the present study, to evaluate whether the gut-brain-liver axis, an important neural pathway for the control of hepatic glucose production, is involved in silibinin-regulated glucose homeostasis, the expression of glucagon-like peptide-1 receptor (GLP1R) in the duodenum, activation of neurons in the nucleus of the solitary tract (NTS), as well as glycogen accumulation and expression of gluconeogenic enzymes in the livers of diabetic SHRSP·Z-Leprfa/IzmDmcr (SP·ZF) rats with 4-week oral administration of silibinin (100 and 300 mg kg-1 day-1) were evaluated. Common hepatic branch vagotomy was further conducted in high-fat diet/streptozotocin (HFD/STZ)-induced diabetic SD rats to confirm the role of the gut-brain-liver axis in silibinin-improved glycemic control. The results revealed a significant inhibition of fasting blood glucose after SP·ZF rats were administrated with silibinin for 4 weeks. The expression of GLP1R in the duodenum and the activation of neurons in the NTS increased, while hepatic glucose production decreased on silibinin administration. However, the hypoglycemic effect of silibinin was reversed by common hepatic branch vagotomy in diabetic SD rats. Our study suggested that silibinin may be useful as a potential functional food ingredient against diabetes by triggering the gut-brain-liver axis.
Collapse
Affiliation(s)
- Fanxing Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, P.R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
119
|
Niccolai E, Boem F, Russo E, Amedei A. The Gut⁻Brain Axis in the Neuropsychological Disease Model of Obesity: A Classical Movie Revised by the Emerging Director "Microbiome". Nutrients 2019; 11:156. [PMID: 30642052 PMCID: PMC6356219 DOI: 10.3390/nu11010156] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/21/2018] [Accepted: 01/08/2019] [Indexed: 02/06/2023] Open
Abstract
The worldwide epidemic of obesity has become an important public health issue, with serious psychological and social consequences. Obesity is a multifactorial disorder in which various elements (genetic, host, and environment), play a definite role, even if none of them satisfactorily explains its etiology. A number of neurological comorbidities, such as anxiety and depression, charges the global obesity burden, and evidence suggests the hypothesis that the brain could be the seat of the initial malfunction leading to obesity. The gut microbiome plays an important role in energy homeostasis regulating energy harvesting, fat deposition, as well as feeding behavior and appetite. Dietary patterns, like the Western diet, are known to be a major cause of the obesity epidemic, probably promoting a dysbiotic drift in the gut microbiota. Moreover, the existence of a "gut⁻brain axis" suggests a role for microbiome on hosts' behavior according to different modalities, including interaction through the nervous system, and mutual crosstalk with the immune and the endocrine systems. In the perspective of obesity as a real neuropsychological disease and in light of the discussed considerations, this review focuses on the microbiome role as an emerging director in the development of obesity.
Collapse
Affiliation(s)
- Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.
| | - Federico Boem
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.
| | - Edda Russo
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.
- Department of Biomedicine, Azienda Ospedaliera Universitaria Careggi (AOUC), Largo Brambilla 3, 50134 Florence, Italy.
| |
Collapse
|
120
|
Yahaya TO, Salisu TF. A Review of Type 2 Diabetes Mellitus Predisposing Genes. Curr Diabetes Rev 2019; 16:52-61. [PMID: 30514191 DOI: 10.2174/1573399815666181204145806] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 11/22/2018] [Accepted: 11/28/2018] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Scientists are considering the possibility of treating diabetes mellitus (DM) using a personalized approach in which various forms of the diseases will be treated based on the causal gene and its pathogenesis. To this end, scientists have identified mutations in certain genes as probable causes of Type 2 diabetes mellitus (T2DM) with diverse mechanisms. AIM This review was aimed at articulating already identified T2DM genes with their mechanisms of action and phenotypic presentations for the awareness of all stakeholders. METHOD The Google search engine was used to retrieve relevant information on the subject from reliable academic databases such as PubMed, Medline, and Google Scholar, among others. RESULTS At least seventy (70) genes are currently being suspected in the biogenesis of T2DM. However, mutations in, or variants of KCNJ11, PPARG, HNF1B and WFS1 genes, are the most suspected and reported in the pathogenesis of the disease. Mutations in these genes can cause disruption of insulin biosynthesis through the destruction of pancreatic beta cells, change of beta cell morphology, destruction of insulin receptors, among others. These cellular events may lead to insulin resistance and hyperglycemia and, along with environmental triggers such as obesity and overweight, culminate in T2DM. It was observed that each identified gene has its distinct mechanism by which it interacts with other genes and environmental factors to cause T2DM. CONCLUSION Healthcare providers are advised to formulate T2DM drugs or treatment by targeting the causal genes along with their mechanisms.
Collapse
Affiliation(s)
- Tajudeen O Yahaya
- Department of Biology, Federal University Birnin Kebbi, Kebbi State, Nigeria
| | - Titilola F Salisu
- Department of Cell Biology and Genetics, University of Lagos, Lagos, Nigeria
| |
Collapse
|
121
|
Balland E, Chen W, Dodd GT, Conductier G, Coppari R, Tiganis T, Cowley MA. Leptin Signaling in the Arcuate Nucleus Reduces Insulin’s Capacity to Suppress Hepatic Glucose Production in Obese Mice. Cell Rep 2019; 26:346-355.e3. [DOI: 10.1016/j.celrep.2018.12.061] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 11/29/2018] [Accepted: 12/13/2018] [Indexed: 12/18/2022] Open
|
122
|
Njan AA, Fatigun CO, Alli-Oluwafuyi AM, Olorundare OE, Afolabi OS, Akinola O, Amin A. Effect of intranasal insulin on peripheral glucose profile in dexamethasone-induced insulin resistance in Wistar rats. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2018. [DOI: 10.1016/j.bjbas.2018.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
|
123
|
Santoleri D, Titchenell PM. Resolving the Paradox of Hepatic Insulin Resistance. Cell Mol Gastroenterol Hepatol 2018; 7:447-456. [PMID: 30739869 PMCID: PMC6369222 DOI: 10.1016/j.jcmgh.2018.10.016] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 10/30/2018] [Accepted: 10/30/2018] [Indexed: 12/12/2022]
Abstract
Insulin resistance is associated with numerous metabolic disorders, such as obesity and type II diabetes, that currently plague our society. Although insulin normally promotes anabolic metabolism in the liver by increasing glucose consumption and lipid synthesis, insulin-resistant individuals fail to inhibit hepatic glucose production and paradoxically have increased liver lipid synthesis, leading to hyperglycemia and hypertriglyceridemia. Here, we detail the intrahepatic and extrahepatic pathways mediating insulin's control of glucose and lipid metabolism. We propose that the interplay between both of these pathways controls insulin signaling and that mis-regulation between the 2 results in the paradoxic effects seen in the insulin-resistant liver instead of the commonly proposed deficiencies in particular branches of only the direct hepatic pathway.
Collapse
Affiliation(s)
- Dominic Santoleri
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Paul M. Titchenell
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania,Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania,Correspondence Address correspondence to: Paul M. Titchenell, PhD, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, Pennsylvania 19104. fax: (215) 898-5408.
| |
Collapse
|
124
|
Metz CN, Pavlov VA. Vagus nerve cholinergic circuitry to the liver and the gastrointestinal tract in the neuroimmune communicatome. Am J Physiol Gastrointest Liver Physiol 2018; 315:G651-G658. [PMID: 30001146 PMCID: PMC6293249 DOI: 10.1152/ajpgi.00195.2018] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Improved understanding of neuroimmune communication and the neural regulation of immunity and inflammation has recently led to proposing the concept of the "neuroimmune communicatome." This advance is based on experimental evidence for an organized and brain-integrated reflex-like relationship and dialogue between the nervous and the immune systems. A key circuitry in this communicatome is provided by efferent vagus nerve fibers and cholinergic signaling. Inflammation and metabolic alterations coexist in many disorders affecting the liver and the gastrointestinal (GI) tract, including obesity, metabolic syndrome, fatty liver disease, liver injury, and liver failure, as well as inflammatory bowel disease. Here, we outline mechanistic insights regarding the role of the vagus nerve and cholinergic signaling in the regulation of inflammation linked to metabolic derangements and the pathogenesis of these disorders in preclinical settings. Recent clinical advances using this knowledge in novel therapeutic neuromodulatory approaches within the field of bioelectronic medicine are also briefly summarized.
Collapse
Affiliation(s)
- Christine N. Metz
- 1Center for Biomedical Science and Bioelectronic Medicine, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York,2Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
| | - Valentin A. Pavlov
- 1Center for Biomedical Science and Bioelectronic Medicine, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York,2Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
| |
Collapse
|
125
|
Petersen MC, Shulman GI. Mechanisms of Insulin Action and Insulin Resistance. Physiol Rev 2018; 98:2133-2223. [PMID: 30067154 PMCID: PMC6170977 DOI: 10.1152/physrev.00063.2017] [Citation(s) in RCA: 1701] [Impact Index Per Article: 243.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/22/2018] [Accepted: 03/24/2018] [Indexed: 12/15/2022] Open
Abstract
The 1921 discovery of insulin was a Big Bang from which a vast and expanding universe of research into insulin action and resistance has issued. In the intervening century, some discoveries have matured, coalescing into solid and fertile ground for clinical application; others remain incompletely investigated and scientifically controversial. Here, we attempt to synthesize this work to guide further mechanistic investigation and to inform the development of novel therapies for type 2 diabetes (T2D). The rational development of such therapies necessitates detailed knowledge of one of the key pathophysiological processes involved in T2D: insulin resistance. Understanding insulin resistance, in turn, requires knowledge of normal insulin action. In this review, both the physiology of insulin action and the pathophysiology of insulin resistance are described, focusing on three key insulin target tissues: skeletal muscle, liver, and white adipose tissue. We aim to develop an integrated physiological perspective, placing the intricate signaling effectors that carry out the cell-autonomous response to insulin in the context of the tissue-specific functions that generate the coordinated organismal response. First, in section II, the effectors and effects of direct, cell-autonomous insulin action in muscle, liver, and white adipose tissue are reviewed, beginning at the insulin receptor and working downstream. Section III considers the critical and underappreciated role of tissue crosstalk in whole body insulin action, especially the essential interaction between adipose lipolysis and hepatic gluconeogenesis. The pathophysiology of insulin resistance is then described in section IV. Special attention is given to which signaling pathways and functions become insulin resistant in the setting of chronic overnutrition, and an alternative explanation for the phenomenon of ‟selective hepatic insulin resistanceˮ is presented. Sections V, VI, and VII critically examine the evidence for and against several putative mediators of insulin resistance. Section V reviews work linking the bioactive lipids diacylglycerol, ceramide, and acylcarnitine to insulin resistance; section VI considers the impact of nutrient stresses in the endoplasmic reticulum and mitochondria on insulin resistance; and section VII discusses non-cell autonomous factors proposed to induce insulin resistance, including inflammatory mediators, branched-chain amino acids, adipokines, and hepatokines. Finally, in section VIII, we propose an integrated model of insulin resistance that links these mediators to final common pathways of metabolite-driven gluconeogenesis and ectopic lipid accumulation.
Collapse
Affiliation(s)
- Max C Petersen
- Departments of Internal Medicine and Cellular & Molecular Physiology, Howard Hughes Medical Institute, Yale University School of Medicine , New Haven, Connecticut
| | - Gerald I Shulman
- Departments of Internal Medicine and Cellular & Molecular Physiology, Howard Hughes Medical Institute, Yale University School of Medicine , New Haven, Connecticut
| |
Collapse
|
126
|
Ribeiro IMR, Antunes VR. The role of insulin at brain-liver axis in the control of glucose production. Am J Physiol Gastrointest Liver Physiol 2018; 315:G538-G543. [PMID: 29878846 DOI: 10.1152/ajpgi.00290.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Glucose is an essential metabolic substrate for all mammalian cells, and its availability in the circulation is carefully controlled to avoid wide variations. Different mechanisms are involved in the glucose disposal, such as an adequate pancreatic and hepatic function. Insulin is the main hormone in glycemic control, and its action occurs directly in the cells, as well as in the liver, in an indirect way, to ultimately control the glycemia. Insulin has also an important action within the central nervous system, more precisely in the hypothalamus that projects directly to preautonomic nuclei in the brain stem to control hepatic glucose production. The central action of insulin relies on autonomic outflow through the vagal innervation of the liver, where insulin is able to modulate the production of glucose at this organ level. In this way, responses generated in the CNS reach the effector organs by autonomic efferent pathways as part of an important brain-organ axis in the control of glycemia. The purpose of this minireview is to shed light on the brain-liver axis in the control of hepatic glucose by central action of insulin via the autonomic nervous system.
Collapse
Affiliation(s)
- Izabela Martina Ramos Ribeiro
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , Brazil
| | - Vagner Roberto Antunes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , Brazil
| |
Collapse
|
127
|
Dodd GT, Michael NJ, Lee-Young RS, Mangiafico SP, Pryor JT, Munder AC, Simonds SE, Brüning JC, Zhang ZY, Cowley MA, Andrikopoulos S, Horvath TL, Spanswick D, Tiganis T. Insulin regulates POMC neuronal plasticity to control glucose metabolism. eLife 2018; 7:38704. [PMID: 30230471 PMCID: PMC6170188 DOI: 10.7554/elife.38704] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 09/14/2018] [Indexed: 11/30/2022] Open
Abstract
Hypothalamic neurons respond to nutritional cues by altering gene expression and neuronal excitability. The mechanisms that control such adaptive processes remain unclear. Here we define populations of POMC neurons in mice that are activated or inhibited by insulin and thereby repress or inhibit hepatic glucose production (HGP). The proportion of POMC neurons activated by insulin was dependent on the regulation of insulin receptor signaling by the phosphatase TCPTP, which is increased by fasting, degraded after feeding and elevated in diet-induced obesity. TCPTP-deficiency enhanced insulin signaling and the proportion of POMC neurons activated by insulin to repress HGP. Elevated TCPTP in POMC neurons in obesity and/or after fasting repressed insulin signaling, the activation of POMC neurons by insulin and the insulin-induced and POMC-mediated repression of HGP. Our findings define a molecular mechanism for integrating POMC neural responses with feeding to control glucose metabolism.
Collapse
Affiliation(s)
- Garron T Dodd
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Natalie J Michael
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia.,Department of Physiology, Monash University, Victoria, Australia
| | - Robert S Lee-Young
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia.,Monash Metabolic Phenotyping Facility, Monash University, Victoria, Australia
| | - Salvatore P Mangiafico
- Department of Medicine (Austin Hospital), The University of Melbourne, Melbourne, Australia
| | - Jack T Pryor
- Department of Physiology, Monash University, Victoria, Australia.,Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Astrid C Munder
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia.,Department of Physiology, Monash University, Victoria, Australia
| | - Stephanie E Simonds
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia.,Department of Physiology, Monash University, Victoria, Australia
| | - Jens Claus Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany.,Center for Endocrinology, Diabetes, and Preventive Medicine, University Hospital Cologne, Cologne, Germany.,Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,National Center for Diabetes Research, Neuherberg, Germany
| | - Zhong-Yin Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, United States
| | - Michael A Cowley
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia.,Department of Physiology, Monash University, Victoria, Australia
| | - Sofianos Andrikopoulos
- Department of Medicine (Austin Hospital), The University of Melbourne, Melbourne, Australia
| | - Tamas L Horvath
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, United States.,Department of Anatomy and Histology, University of Veterinary Medicine, Hungary, Europe
| | - David Spanswick
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia.,Department of Physiology, Monash University, Victoria, Australia.,Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Tony Tiganis
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia.,Monash Metabolic Phenotyping Facility, Monash University, Victoria, Australia
| |
Collapse
|
128
|
Ma Y, Ratnasabapathy R, Izzi-Engbeaya C, Nguyen-Tu MS, Richardson E, Hussain S, De Backer I, Holton C, Norton M, Carrat G, Schwappach B, Rutter GA, Dhillo WS, Gardiner J. Hypothalamic arcuate nucleus glucokinase regulates insulin secretion and glucose homeostasis. Diabetes Obes Metab 2018; 20:2246-2254. [PMID: 29748994 PMCID: PMC6099255 DOI: 10.1111/dom.13359] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 04/30/2018] [Accepted: 05/09/2018] [Indexed: 01/08/2023]
Abstract
AIMS To investigate the role of arcuate glucokinase (GK) in the regulation of glucose homeostasis. MATERIALS AND METHODS A recombinant adeno-associated virus expressing either GK or an antisense GK construct was used to alter GK activity specifically in the hypothalamic arcuate nucleus (arc). GK activity in this nucleus was also increased by stereotactic injection of the GK activator, compound A. The effect of altered arc GK activity on glucose homeostasis was subsequently investigated using glucose and insulin tolerance tests. RESULTS Increased GK activity specifically within the arc increased insulin secretion and improved glucose tolerance in rats during oral glucose tolerance tests. Decreased GK activity in this nucleus reduced insulin secretion and increased glucose levels during the same tests. Insulin sensitivity was not affected in either case. The effect of arc GK was maintained in a model of type 2 diabetes. CONCLUSIONS These results demonstrate a role for arc GK in systemic glucose homeostasis.
Collapse
Affiliation(s)
- Yue Ma
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Risheka Ratnasabapathy
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Chioma Izzi-Engbeaya
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Marie-Sophie Nguyen-Tu
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Errol Richardson
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Sufyan Hussain
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Ivan De Backer
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Christopher Holton
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Mariana Norton
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Gaelle Carrat
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Blanche Schwappach
- Department of Molecular Biology, Centre for Biochemistry and Molecular Cell Biology, Heart Research Centre Göttingen, University Medicine Göttingen, Göttingen, Germany
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Waljit S Dhillo
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - James Gardiner
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| |
Collapse
|
129
|
Huang X, Liu G, Guo J, Su Z. The PI3K/AKT pathway in obesity and type 2 diabetes. Int J Biol Sci 2018; 14:1483-1496. [PMID: 30263000 PMCID: PMC6158718 DOI: 10.7150/ijbs.27173] [Citation(s) in RCA: 987] [Impact Index Per Article: 141.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 07/10/2018] [Indexed: 02/06/2023] Open
Abstract
Obesity and type 2 diabetes mellitus are complicated metabolic diseases that affect multiple organs and are characterized by hyperglycaemia. Currently, stable and effective treatments for obesity and type 2 diabetes mellitus are not available. Therefore, the mechanisms leading to obesity and diabetes and more effective ways to treat obesity and diabetes should be identified. Based on accumulated evidences, the PI3K/AKT signalling pathway is required for normal metabolism due to its characteristics, and its imbalance leads to the development of obesity and type 2 diabetes mellitus. This review focuses on the role of PI3K/AKT signalling in the skeletal muscle, adipose tissue, liver, brain and pancreas, and discusses how this signalling pathway affects the development of the aforementioned diseases. We also summarize evidences for recently identified therapeutic targets of the PI3K/AKT pathway as treatments for obesity and type 2 diabetes mellitus. PI3K/AKT pathway damaged in various tissues of the body leads to obesity and type 2 diabetes as the result of insulin resistance, and in turn, insulin resistance exacerbates the PI3K/AKT pathway, forming a vicious circle.
Collapse
Affiliation(s)
- Xingjun Huang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou (510006), China.,Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou (510006), China
| | - Guihua Liu
- Shenzhen Center for Disease Control and Prevention, 8 Longyuan Road, Nanshan District, Shenzhen (518055), China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou (510006), China
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou (510006), China.,Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou (510006), China
| |
Collapse
|
130
|
Onyango AN. Cellular Stresses and Stress Responses in the Pathogenesis of Insulin Resistance. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4321714. [PMID: 30116482 PMCID: PMC6079365 DOI: 10.1155/2018/4321714] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 02/18/2018] [Indexed: 12/14/2022]
Abstract
Insulin resistance (IR), a key component of the metabolic syndrome, precedes the development of diabetes, cardiovascular disease, and Alzheimer's disease. Its etiological pathways are not well defined, although many contributory mechanisms have been established. This article summarizes such mechanisms into the hypothesis that factors like nutrient overload, physical inactivity, hypoxia, psychological stress, and environmental pollutants induce a network of cellular stresses, stress responses, and stress response dysregulations that jointly inhibit insulin signaling in insulin target cells including endothelial cells, hepatocytes, myocytes, hypothalamic neurons, and adipocytes. The insulin resistance-inducing cellular stresses include oxidative, nitrosative, carbonyl/electrophilic, genotoxic, and endoplasmic reticulum stresses; the stress responses include the ubiquitin-proteasome pathway, the DNA damage response, the unfolded protein response, apoptosis, inflammasome activation, and pyroptosis, while the dysregulated responses include the heat shock response, autophagy, and nuclear factor erythroid-2-related factor 2 signaling. Insulin target cells also produce metabolites that exacerbate cellular stress generation both locally and systemically, partly through recruitment and activation of myeloid cells which sustain a state of chronic inflammation. Thus, insulin resistance may be prevented or attenuated by multiple approaches targeting the different cellular stresses and stress responses.
Collapse
Affiliation(s)
- Arnold N. Onyango
- Department of Food Science and Technology, Jomo Kenyatta University of Agriculture and Technology, P.O. Box 62000, Nairobi 00200, Kenya
| |
Collapse
|
131
|
Güemes A, Georgiou P. Review of the role of the nervous system in glucose homoeostasis and future perspectives towards the management of diabetes. Bioelectron Med 2018; 4:9. [PMID: 32232085 PMCID: PMC7098234 DOI: 10.1186/s42234-018-0009-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/10/2018] [Indexed: 12/16/2022] Open
Abstract
Diabetes is a disease caused by a breakdown in the glucose metabolic process resulting in abnormal blood glucose fluctuations. Traditionally, control has involved external insulin injection in response to elevated blood glucose to substitute the role of the beta cells in the pancreas which would otherwise perform this function in a healthy individual. The central nervous system (CNS), however, also plays a vital role in glucose homoeostasis through the control of pancreatic secretion and insulin sensitivity which could potentially be used as a pathway for enhancing glucose control. In this review, we present an overview of the brain regions, peripheral nerves and molecular mechanisms by which the CNS regulates glucose metabolism and the potential benefits of modulating them for diabetes management. Development of technologies to interface to the nervous system will soon become a reality through bioelectronic medicine and we present the emerging opportunities for the treatment of type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Amparo Güemes
- Centre for Bio-Inspired Technology, Department of Electrical and Electronic Engineering, Imperial College London, South Kensington Campus, London, UK
| | - Pantelis Georgiou
- Centre for Bio-Inspired Technology, Department of Electrical and Electronic Engineering, Imperial College London, South Kensington Campus, London, UK
| |
Collapse
|
132
|
Dodd GT, Lee-Young RS, Brüning JC, Tiganis T. TCPTP Regulates Insulin Signaling in AgRP Neurons to Coordinate Glucose Metabolism With Feeding. Diabetes 2018; 67:1246-1257. [PMID: 29712668 DOI: 10.2337/db17-1485] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 04/19/2018] [Indexed: 11/13/2022]
Abstract
Insulin regulates glucose metabolism by eliciting effects on peripheral tissues as well as the brain. Insulin receptor (IR) signaling inhibits AgRP-expressing neurons in the hypothalamus to contribute to the suppression of hepatic glucose production (HGP) by insulin, whereas AgRP neuronal activation attenuates brown adipose tissue (BAT) glucose uptake. The tyrosine phosphatase TCPTP suppresses IR signaling in AgRP neurons. Hypothalamic TCPTP is induced by fasting and degraded after feeding. Here we assessed the influence of TCPTP in AgRP neurons in the control of glucose metabolism. TCPTP deletion in AgRP neurons (Agrp-Cre;Ptpn2fl/fl ) enhanced insulin sensitivity, as assessed by the increased glucose infusion rates, and reduced HGP during hyperinsulinemic-euglycemic clamps, accompanied by increased [14C]-2-deoxy-d-glucose uptake in BAT and browned white adipose tissue. TCPTP deficiency in AgRP neurons promoted the intracerebroventricular insulin-induced repression of hepatic gluconeogenesis in otherwise unresponsive food-restricted mice, yet had no effect in fed/satiated mice where hypothalamic TCPTP levels are reduced. The improvement in glucose homeostasis in Agrp-Cre;Ptpn2fl/fl mice was corrected by IR heterozygosity (Agrp-Cre;Ptpn2fl/fl ;Insrfl/+ ), causally linking the effects on glucose metabolism with the IR signaling in AgRP neurons. Our findings demonstrate that TCPTP controls IR signaling in AgRP neurons to coordinate HGP and brown/beige adipocyte glucose uptake in response to feeding/fasting.
Collapse
Affiliation(s)
- Garron T Dodd
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Melbourne, Victoria, Australia
| | - Robert S Lee-Young
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Melbourne, Victoria, Australia
- Monash Metabolic Phenotyping Facility, Monash University, Clayton, Melbourne, Victoria, Australia
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Plank Institute for Metabolism Research, Cologne, Germany
- Center for Endocrinology, Diabetes, and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- National Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Tony Tiganis
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Melbourne, Victoria, Australia
- Monash Metabolic Phenotyping Facility, Monash University, Clayton, Melbourne, Victoria, Australia
| |
Collapse
|
133
|
Page MM, Johnson JD. Mild Suppression of Hyperinsulinemia to Treat Obesity and Insulin Resistance. Trends Endocrinol Metab 2018; 29:389-399. [PMID: 29665988 DOI: 10.1016/j.tem.2018.03.018] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/23/2018] [Accepted: 03/27/2018] [Indexed: 12/14/2022]
Abstract
Insulin plays roles in lipid uptake, lipolysis, and lipogenesis, in addition to controlling blood glucose levels. Excessive circulating insulin is associated with adipose tissue expansion and obesity, yet a causal role for hyperinsulinemia in the development of mammalian obesity has proven controversial, with many researchers suggesting it as a consequence of insulin resistance. Recently, evidence that specifically reducing hyperinsulinemia can prevent and reverse obesity in animal models has been presented. Our experiments, and others in this field, question the current dogma that hyperinsulinemia is a response to obesity and/or insulin resistance. In this review, we discuss preclinical evidence in the context of the broader literature and speculate on the possibility of clinical translation of alternative approaches for treating obesity.
Collapse
Affiliation(s)
- Melissa M Page
- Life Sciences Institute Diabetes Research Group and the Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada; Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - James D Johnson
- Life Sciences Institute Diabetes Research Group and the Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada. https://twitter.com/JimJohnsonSci
| |
Collapse
|
134
|
Abstract
While there is a growing consensus that insulin has diverse and important regulatory actions on the brain, seemingly important aspects of brain insulin physiology are poorly understood. Examples include: what is the insulin concentration within brain interstitial fluid under normal physiologic conditions; whether insulin is made in the brain and acts locally; does insulin from the circulation cross the blood-brain barrier or the blood-CSF barrier in a fashion that facilitates its signaling in brain; is insulin degraded within the brain; do privileged areas with a "leaky" blood-brain barrier serve as signaling nodes for transmitting peripheral insulin signaling; does insulin action in the brain include regulation of amyloid peptides; whether insulin resistance is a cause or consequence of processes involved in cognitive decline. Heretofore, nearly all of the studies examining brain insulin physiology have employed techniques and methodologies that do not appreciate the complex fluid compartmentation and flow throughout the brain. This review attempts to provide a status report on historical and recent work that begins to address some of these issues. It is undertaken in an effort to suggest a framework for studies going forward. Such studies are inevitably influenced by recent physiologic and genetic studies of insulin accessing and acting in brain, discoveries relating to brain fluid dynamics and the interplay of cerebrospinal fluid, brain interstitial fluid, and brain lymphatics, and advances in clinical neuroimaging that underscore the dynamic role of neurovascular coupling.
Collapse
Affiliation(s)
- Sarah M Gray
- Department of Pharmacology, Department of Medicine, University of Virginia, School of Medicine , Charlottesville, Virginia
| | - Eugene J Barrett
- Department of Pharmacology, Department of Medicine, University of Virginia, School of Medicine , Charlottesville, Virginia.,Division of Endocrinology, Department of Medicine, University of Virginia, School of Medicine , Charlottesville, Virginia
| |
Collapse
|
135
|
Yin Z, Raj DD, Schaafsma W, van der Heijden RA, Kooistra SM, Reijne AC, Zhang X, Moser J, Brouwer N, Heeringa P, Yi CX, van Dijk G, Laman JD, Boddeke EWGM, Eggen BJL. Low-Fat Diet With Caloric Restriction Reduces White Matter Microglia Activation During Aging. Front Mol Neurosci 2018; 11:65. [PMID: 29593493 PMCID: PMC5857900 DOI: 10.3389/fnmol.2018.00065] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 02/15/2018] [Indexed: 12/31/2022] Open
Abstract
Rodent models of both aging and obesity are characterized by inflammation in specific brain regions, notably the corpus callosum, fornix, and hypothalamus. Microglia, the resident macrophages of the central nervous system, are important for brain development, neural support, and homeostasis. However, the effects of diet and lifestyle on microglia during aging are only partly understood. Here, we report alterations in microglia phenotype and functions in different brain regions of mice on a high-fat diet (HFD) or low-fat diet (LFD) during aging and in response to voluntary running wheel exercise. We compared the expression levels of genes involved in immune response, phagocytosis, and metabolism in the hypothalamus of 6-month-old HFD and LFD mice. We also compared the immune response of microglia from HFD or LFD mice to peripheral inflammation induced by intraperitoneal injection of lipopolysaccharide (LPS). Finally, we investigated the effect of diet, physical exercise, and caloric restriction (40% reduction compared to ad libitum intake) on microglia in 24-month-old HFD and LFD mice. Changes in diet caused morphological changes in microglia, but did not change the microglia response to LPS-induced systemic inflammation. Expression of phagocytic markers (i.e., Mac-2/Lgals3, Dectin-1/Clec7a, and CD16/CD32) in the white matter microglia of 24-month-old brain was markedly decreased in calorically restricted LFD mice. In conclusion, LFD resulted in reduced activation of microglia, which might be an underlying mechanism for the protective role of caloric restriction during aging-associated decline.
Collapse
Affiliation(s)
- Zhuoran Yin
- Department of Neurology, Tongji Hospital, Tongji Medical College of HUST, Huazhong University of Science and Technology, Wuhan, China
- Department of Neuroscience, Medical Physiology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Divya D. Raj
- Department of Neuroscience, Medical Physiology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Wandert Schaafsma
- Department of Neuroscience, Medical Physiology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Roel A. van der Heijden
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Susanne M. Kooistra
- Department of Neuroscience, Medical Physiology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Aaffien C. Reijne
- Laboratory of Pediatrics, Systems Medicine of Metabolism and Signaling Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Systems Biology Centre for Energy Metabolism and Ageing, University of Groningen, Groningen, Netherlands
- Groningen Institute for Evolutionary Life Sciences, Department of Behavioral Neuroscience, University of Groningen, Groningen, Netherlands
| | - Xiaoming Zhang
- Department of Neuroscience, Medical Physiology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jill Moser
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Nieske Brouwer
- Department of Neuroscience, Medical Physiology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Peter Heeringa
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Gertjan van Dijk
- Systems Biology Centre for Energy Metabolism and Ageing, University of Groningen, Groningen, Netherlands
- Groningen Institute for Evolutionary Life Sciences, Department of Behavioral Neuroscience, University of Groningen, Groningen, Netherlands
- ESRIG Centre for Isotope Research, University of Groningen, Groningen, Netherlands
| | - Jon D. Laman
- Department of Neuroscience, Medical Physiology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Erik W. G. M. Boddeke
- Department of Neuroscience, Medical Physiology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Bart J. L. Eggen
- Department of Neuroscience, Medical Physiology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
136
|
Brain insulin resistance in type 2 diabetes and Alzheimer disease: concepts and conundrums. Nat Rev Neurol 2018; 14:168-181. [PMID: 29377010 DOI: 10.1038/nrneurol.2017.185] [Citation(s) in RCA: 1002] [Impact Index Per Article: 143.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Considerable overlap has been identified in the risk factors, comorbidities and putative pathophysiological mechanisms of Alzheimer disease and related dementias (ADRDs) and type 2 diabetes mellitus (T2DM), two of the most pressing epidemics of our time. Much is known about the biology of each condition, but whether T2DM and ADRDs are parallel phenomena arising from coincidental roots in ageing or synergistic diseases linked by vicious pathophysiological cycles remains unclear. Insulin resistance is a core feature of T2DM and is emerging as a potentially important feature of ADRDs. Here, we review key observations and experimental data on insulin signalling in the brain, highlighting its actions in neurons and glia. In addition, we define the concept of 'brain insulin resistance' and review the growing, although still inconsistent, literature concerning cognitive impairment and neuropathological abnormalities in T2DM, obesity and insulin resistance. Lastly, we review evidence of intrinsic brain insulin resistance in ADRDs. By expanding our understanding of the overlapping mechanisms of these conditions, we hope to accelerate the rational development of preventive, disease-modifying and symptomatic treatments for cognitive dysfunction in T2DM and ADRDs alike.
Collapse
|
137
|
Page MM, Skovsø S, Cen H, Chiu AP, Dionne DA, Hutchinson DF, Lim GE, Szabat M, Flibotte S, Sinha S, Nislow C, Rodrigues B, Johnson JD. Reducing insulin via conditional partial gene ablation in adults reverses diet-induced weight gain. FASEB J 2018; 32:1196-1206. [PMID: 29122848 PMCID: PMC5892722 DOI: 10.1096/fj.201700518r] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Excess circulating insulin is associated with obesity in humans and in animal models. However, the physiologic causality of hyperinsulinemia in adult obesity has rightfully been questioned because of the absence of clear evidence that weight loss can be induced by acutely reversing diet-induced hyperinsulinemia. Herein, we describe the consequences of inducible, partial insulin gene deletion in a mouse model in which animals have already been made obese by consuming a high-fat diet. A modest reduction in insulin production/secretion was sufficient to cause significant weight loss within 5 wk, with a specific effect on visceral adipose tissue. This result was associated with a reduction in the protein abundance of the lipodystrophy gene polymerase I and transcript release factor ( Ptrf; Cavin) in gonadal adipose tissue. RNAseq analysis showed that reduced insulin and weight loss also associated with a signature of reduced innate immunity. This study demonstrates that changes in circulating insulin that are too fine to adversely affect glucose homeostasis nonetheless exert control over adiposity.-Page, M. M., Skovsø, S., Cen, H., Chiu, A. P., Dionne, D. A., Hutchinson, D. F., Lim, G. E., Szabat, M., Flibotte, S., Sinha, S., Nislow, C., Rodrigues, B., Johnson, J. D. Reducing insulin via conditional partial gene ablation in adults reverses diet-induced weight gain.
Collapse
Affiliation(s)
- Melissa M Page
- Department of Cellular and Physiological Sciences, Diabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Søs Skovsø
- Department of Cellular and Physiological Sciences, Diabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Haoning Cen
- Department of Cellular and Physiological Sciences, Diabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amy P Chiu
- Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Derek A Dionne
- Department of Cellular and Physiological Sciences, Diabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Daria F Hutchinson
- Department of Cellular and Physiological Sciences, Diabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gareth E Lim
- Department of Cellular and Physiological Sciences, Diabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Marta Szabat
- Department of Cellular and Physiological Sciences, Diabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephane Flibotte
- Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sunita Sinha
- Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Corey Nislow
- Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Brian Rodrigues
- Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - James D Johnson
- Department of Cellular and Physiological Sciences, Diabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
138
|
Rodriguez-Raecke R, Brünner YF, Kofoet A, Mutic S, Benedict C, Freiherr J. Odor Sensitivity After Intranasal Insulin Application Is Modulated by Gender. Front Endocrinol (Lausanne) 2018; 9:580. [PMID: 30356884 PMCID: PMC6190874 DOI: 10.3389/fendo.2018.00580] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 09/12/2018] [Indexed: 11/13/2022] Open
Abstract
Obesity constitutes a global health care problem, and often eating habits are to blame. For intervention, a thorough understanding of energy intake and expenditure is needed. In recent years, the pivotal role of insulin in connection to energy intake was established. Olfactory sensitivity may be a target of cerebral insulin action to maintain body weight. With this experiment, we aimed to explore the influence of intranasal insulin on olfactory sensitivity for the odors n-butanol and peanut in a placebo-controlled, double-blind setting in a within-subject design. All subjects participated in two experimental sessions on separate days and received either intranasal insulin or placebo in a pseudorandomized order. Application was followed by two olfactory threshold tests for n-butanol and peanut in a pseudorandomized order. After a single dose of intranasal insulin (40 IU) or placebo (0.4 ml), olfactory sensitivity for the odorants n-butanol and peanut were examined in 30 healthy normosmic participants (14 females). Measured blood parameters revealed no decrease in plasma glucose, however, insulin, leptin and cortisol levels were affected following intranasal application. Females' but not males' olfactory sensitivity for n-butanol was lower after intranasal insulin administration vs. placebo. In contrast, olfactory sensitivity for peanut was not influenced by intranasal insulin application. Our results indicate that the effects of cortical insulin levels on processing of specific odors is likely modulated by gender, as central increase of insulin concentration led to a reduced olfactory sensitivity for n-butanol in women only, which might be due to differentially regulated insulin and leptin signaling in men and women.
Collapse
Affiliation(s)
- Rea Rodriguez-Raecke
- Diagnostic and Interventional Neuroradiology, University Hospital, RWTH Aachen University, Aachen, Germany
- Sensory Analytics, Fraunhofer Institute for Process Engineering and Packaging IVV, Freising, Germany
- *Correspondence: Rea Rodriguez-Raecke
| | - Yvonne F. Brünner
- Diagnostic and Interventional Neuroradiology, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Anja Kofoet
- Diagnostic and Interventional Neuroradiology, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Smiljana Mutic
- Diagnostic and Interventional Neuroradiology, University Hospital, RWTH Aachen University, Aachen, Germany
| | | | - Jessica Freiherr
- Diagnostic and Interventional Neuroradiology, University Hospital, RWTH Aachen University, Aachen, Germany
- Sensory Analytics, Fraunhofer Institute for Process Engineering and Packaging IVV, Freising, Germany
- Jessica Freiherr
| |
Collapse
|
139
|
Kullmann S, Veit R, Peter A, Pohmann R, Scheffler K, Häring HU, Fritsche A, Preissl H, Heni M. Dose-Dependent Effects of Intranasal Insulin on Resting-State Brain Activity. J Clin Endocrinol Metab 2018; 103:253-262. [PMID: 29095982 DOI: 10.1210/jc.2017-01976] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 10/23/2017] [Indexed: 02/05/2023]
Abstract
CONTEXT Insulin action in the human brain influences eating behavior, cognition, and whole-body metabolism. Studies investigating brain insulin rely on intranasal application. OBJECTIVE To investigate effects of three doses of insulin and placebo as nasal sprays on the central and autonomous nervous system and analyze absorption of insulin into the bloodstream. DESIGN, PARTICIPANTS, AND METHODS Nine healthy men received placebo or 40 U, 80 U, and 160 U insulin spray in randomized order. Before and after spray, brain activity was assessed by functional magnetic resonance imaging, and heart rate variability (HRV) was assessed from electrocardiogram. Plasma insulin, C-peptide, and glucose were measured regularly. SETTING General community. RESULTS Nasal insulin administration dose-dependently modulated regional brain activity and the normalized high-frequency component of the HRV. Post hoc analyses revealed that only 160 U insulin showed a considerable difference from placebo. Dose-dependent spillover of nasal insulin into the bloodstream was detected. The brain response was not correlated with this temporary rise in circulating insulin. CONCLUSIONS Nasal insulin dose-dependently modulated regional brain activity with the strongest effects after 160 U. However, this dose was accompanied by a transient increase in circulating insulin concentrations due to a spillover into circulation. Our current results may serve as a basis for future studies with nasal insulin to untangle brain insulin effects in health and disease.
Collapse
Affiliation(s)
- Stephanie Kullmann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Tübingen, Germany
| | - Ralf Veit
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Tübingen, Germany
| | - Andreas Peter
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Tübingen, Germany
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Rolf Pohmann
- High-Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Klaus Scheffler
- High-Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
- Department for Biomedical Magnetic Resonance, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Tübingen, Germany
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- Interfaculty Centre for Pharmacogenomics and Pharma Research at the Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Tübingen, Germany
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Hubert Preissl
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Tübingen, Germany
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- Interfaculty Centre for Pharmacogenomics and Pharma Research at the Eberhard Karls University Tübingen, Tübingen, Germany
- Institute of Pharmaceutical Sciences, Department of Pharmacy and Biochemistry, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Martin Heni
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Tübingen, Germany
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
140
|
Lucio D, Martínez-Ohárriz MC, González-Navarro CJ, Navarro-Herrera D, González-Gaitano G, Radulescu A, Irache JM. Coencapsulation of cyclodextrins into poly(anhydride) nanoparticles to improve the oral administration of glibenclamide. A screening on C. elegans. Colloids Surf B Biointerfaces 2017; 163:64-72. [PMID: 29277019 DOI: 10.1016/j.colsurfb.2017.12.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/16/2017] [Accepted: 12/19/2017] [Indexed: 11/17/2022]
Abstract
This work describes the feasibility of poly(anhydride) nanoparticles as carriers for the oral administration of glibenclamide (GB) as well as the in vivo evaluation of their hypolipidemic effect in a C. elegans model. For this purpose, and in order to increase the GB payload, the drug was encapsulated in nanoparticles in presence of cyclodextrins (either βCD or HPβCD). The optimized nanoparticles displayed a size of about 220 nm and a negative zeta potential (-40 mV), with a drug loading up to 52 μg/mg. Small-angle neutron scattering studies suggested an internal fractal-like structure, based on the repetition of spherical blocks of polymeric units (about 5 nm) grouped to form the nanoparticle. X-ray diffraction study confirmed the absence of crystalline GB molecules due to its dispersion into the nanoparticles, either entrapped in the polymer chains and/or included into cyclodextrin cavities. GB-loaded nanoparticles induced a significant reduction in the fat content of C. elegans. This hypolipidemic effect was slightly higher for the nanoparticles prepared with coencapsulated HPβCD (8.2%) than for those prepared with βCD (7.9%) or in the absence of cyclodextrins (7.0%). In summary, the coencapsulation of cyclodextrins into poly(anhydride) nanoparticles could be an interesting strategy to develop new oral formulations of glibenclamide.
Collapse
Affiliation(s)
- David Lucio
- Department of Chemistry, School of Sciences, University of Navarra, Irunlarrea s/n, Pamplona 31080, Navarra, Spain.
| | | | - Carlos J González-Navarro
- Centre for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Irunlarrea s/n, Pamplona 31080, Navarra, Spain.
| | - David Navarro-Herrera
- Centre for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Irunlarrea s/n, Pamplona 31080, Navarra, Spain.
| | - Gustavo González-Gaitano
- Department of Chemistry, School of Sciences, University of Navarra, Irunlarrea s/n, Pamplona 31080, Navarra, Spain.
| | - Aurel Radulescu
- Jülich Centre for Neutron Science, Heinz Maier-Leibnitz Zentrum, Forschungszentrum Jülich GmbH, 85748 Garching, Germany.
| | - Juan M Irache
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy and Nutrition, University of Navarra, Irunlarrea s/n, Pamplona 31080, Navarra, Spain.
| |
Collapse
|
141
|
Kowalchuk C, Teo C, Wilson V, Chintoh A, Lam L, Agarwal SM, Giacca A, Remington GJ, Hahn MK. In male rats, the ability of central insulin to suppress glucose production is impaired by olanzapine, whereas glucose uptake is left intact. J Psychiatry Neurosci 2017; 42. [PMID: 29083297 PMCID: PMC5662464 DOI: 10.1503/jpn.170092] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Insulin receptors are widely expressed in the brain and may represent a crossroad between metabolic and cognitive disorders. Although antipsychotics, such as olanzapine, are the cornerstone treatment for schizophrenia, they are associated with high rates of type 2 diabetes and lack efficacy for illness-related cognitive deficits. Historically, this risk of diabetes was attributed to the weight gain propensity of antipsychotics, but recent work suggests antipsychotics can have weight-independent diabetogenic effects involving unknown brain-mediated mechanisms. Here, we examined whether antipsychotics disrupt central insulin action, hypothesizing that olanzapine would impair the well-established ability of central insulin to supress hepatic glucose production. METHODS Pancreatic euglycemic clamps were used to measure glucose kinetics alongside a central infusion of insulin or vehicle into the third ventricle. Male rats were pretreated with olanzapine or vehicle per our established model of acute olanzapine-induced peripheral insulin resistance. Groups included (central-peripheral) vehicle-vehicle (n = 11), insulin-vehicle (n = 10), insulin-olanzapine (n = 10) and vehicle-olanzapine (n = 8). RESULTS There were no differences in peripheral glucose or insulin levels. Unexpectedly, we showed that central insulin increased glucose uptake, and this effect was not perturbed by olanzapine. We replicated suppression of glucose production by insulin (clamp relative to basal: 77.9% ± 13.1%, all p < 0.05), an effect abolished by olanzapine (insulin-olanzapine: 7.7% ± 14%). LIMITATIONS This study used only male rats and an acute dose of olanzapine. CONCLUSION To our knowledge, this is the first study suggesting olanzapine may impair central insulin sensing, elucidating a potential mechanism of antipsychotic-induced diabetes and opening avenues of investigation related to domains of schizophrenia psychopathology.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Margaret K. Hahn
- Correspondence to: M.K. Hahn, Centre for Addiction and Mental Health, 250 College St, Toronto ON M5T 1R8;
| |
Collapse
|
142
|
Gancheva S, Bierwagen A, Markgraf DF, Bönhof GJ, Murphy KG, Hatziagelaki E, Lundbom J, Ziegler D, Roden M. Constant hepatic ATP concentrations during prolonged fasting and absence of effects of Cerbomed Nemos ® on parasympathetic tone and hepatic energy metabolism. Mol Metab 2017; 7:71-79. [PMID: 29122559 PMCID: PMC5784324 DOI: 10.1016/j.molmet.2017.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 09/19/2017] [Accepted: 10/01/2017] [Indexed: 02/07/2023] Open
Abstract
Objective Brain insulin-induced improvement in glucose homeostasis has been proposed to be mediated by the parasympathetic nervous system. Non-invasive transcutaneous auricular vagus nerve stimulation (taVNS) activating afferent branches of the vagus nerve may prevent hyperglycemia in diabetes models. We examined the effects of 14-min taVNS vs sham stimulation by Cerbomed Nemos® on glucose metabolism, lipids, and hepatic energy homeostasis in fasted healthy humans (n = 10, age 51 ± 6 yrs, BMI 25.5 ± 2.7 kg/m2). Methods Heart rate variability (HRV), reflecting sympathetic and parasympathetic nerve activity, was measured before, during and after taVNS or sham stimulation. Endogenous glucose production was determined using [6,6-2H2]glucose, and hepatic concentrations of triglycerides (HCL), adenosine triphosphate (ATP), and inorganic phosphate (Pi) were quantified from 1H/31P magnetic resonance spectroscopy at baseline and for 180 min following stimulation. Results taVNS did not affect circulating glucose, free fatty acids, insulin, glucagon, or pancreatic polypeptide. Rates of endogenous glucose production (P = 0.79), hepatic HCL, ATP, and Pi were also not different (P = 0.91, P = 0.48 and P = 0.24) between taVNS or sham stimulation. Hepatic HCL, ATP, and Pi remained constant during prolonged fasting for 3 h. No changes in heart rate or shift in cardiac autonomic function from HRV towards sympathetic or parasympathetic predominance were detected. Conclusion Non-invasive vagus stimulation by Cerbomed Nemos® does not acutely modulate the autonomic tone to the visceral organs and thereby does not affect hepatic glucose and energy metabolism. This technique is therefore unable to mimic brain insulin-mediated effects on peripheral homeostasis in humans. Constant hepatic energy metabolism during prolonged fasting. Vagus stimulation with Cerbomed Nemos® does not alter parasympathetic tone. Cerbomed Nemos® does not modulate hepatic glucose and energy metabolism in humans.
Collapse
Affiliation(s)
- Sofiya Gancheva
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany; German Center of Diabetes Research (DZD e.V.), München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Alessandra Bierwagen
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany; German Center of Diabetes Research (DZD e.V.), München-Neuherberg, Germany
| | - Daniel F Markgraf
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany; German Center of Diabetes Research (DZD e.V.), München-Neuherberg, Germany
| | - Gidon J Bönhof
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany; German Center of Diabetes Research (DZD e.V.), München-Neuherberg, Germany
| | - Kevin G Murphy
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Erifili Hatziagelaki
- 2nd Department of Internal Medicine, Research Institute and Diabetes Center, Athens University, "Attikon" University General Hospital, Athens, Greece
| | - Jesper Lundbom
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany; German Center of Diabetes Research (DZD e.V.), München-Neuherberg, Germany
| | - Dan Ziegler
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany; German Center of Diabetes Research (DZD e.V.), München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany; German Center of Diabetes Research (DZD e.V.), München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany.
| |
Collapse
|
143
|
Abstract
The liver is crucial for the maintenance of normal glucose homeostasis - it produces glucose during fasting and stores glucose postprandially. However, these hepatic processes are dysregulated in type 1 and type 2 diabetes mellitus, and this imbalance contributes to hyperglycaemia in the fasted and postprandial states. Net hepatic glucose production is the summation of glucose fluxes from gluconeogenesis, glycogenolysis, glycogen synthesis, glycolysis and other pathways. In this Review, we discuss the in vivo regulation of these hepatic glucose fluxes. In particular, we highlight the importance of indirect (extrahepatic) control of hepatic gluconeogenesis and direct (hepatic) control of hepatic glycogen metabolism. We also propose a mechanism for the progression of subclinical hepatic insulin resistance to overt fasting hyperglycaemia in type 2 diabetes mellitus. Insights into the control of hepatic gluconeogenesis by metformin and insulin and into the role of lipid-induced hepatic insulin resistance in modifying gluconeogenic and net hepatic glycogen synthetic flux are also discussed. Finally, we consider the therapeutic potential of strategies that target hepatosteatosis, hyperglucagonaemia and adipose lipolysis.
Collapse
Affiliation(s)
- Max C Petersen
- Department of Internal Medicine, Yale School of Medicine
- Department of Cellular &Molecular Physiology, Yale School of Medicine
| | | | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine
- Department of Cellular &Molecular Physiology, Yale School of Medicine
- Howard Hughes Medical Institute, Yale School of Medicine, New Haven, Connecticut 06520, USA
| |
Collapse
|
144
|
van den Top M, Zhao FY, Viriyapong R, Michael NJ, Munder AC, Pryor JT, Renaud LP, Spanswick D. The impact of ageing, fasting and high-fat diet on central and peripheral glucose tolerance and glucose-sensing neural networks in the arcuate nucleus. J Neuroendocrinol 2017; 29. [PMID: 28834571 DOI: 10.1111/jne.12528] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 08/01/2017] [Accepted: 08/17/2017] [Indexed: 12/14/2022]
Abstract
Obesity and ageing are risk factors for diabetes. In the present study, we investigated the effects of ageing, obesity and fasting on central and peripheral glucose tolerance and on glucose-sensing neuronal function in the arcuate nucleus of rats, with a view to providing insight into the central mechanisms regulating glucose homeostasis and how they change or are subject to dysfunction with ageing and obesity. We show that, following a glucose load, central glucose tolerance at the level of the cerebrospinal fluid (CSF) and plasma is significantly reduced in rats maintained on a high-fat diet (HFD). With ageing, up to 2 years, central glucose tolerance was impaired in an age-dependent manner, whereas peripheral glucose tolerance remained unaffected. Ageing-induced peripheral glucose intolerance was improved by a 24-hour fast, whereas central glucose tolerance was not corrected. Pre-wean, immature animals have elevated basal plasma glucose levels and a delayed increase in central glucose levels following peripheral glucose injection compared to mature animals. Electrophysiological recording techniques revealed an energy-status-dependent role for glucose-excited, inhibited and adapting neurones, along with glucose-induced changes in synaptic transmission. We conclude that ageing affects central glucose tolerance, whereas HFD profoundly affects central and peripheral glucose tolerance and, in addition, glucose-sensing neurones adapt function in an energy-status-dependent manner.
Collapse
Affiliation(s)
| | - F-Y Zhao
- NeuroSolutions Ltd, Coventry, UK
| | - R Viriyapong
- Warwick Medical School, University of Warwick, Coventry, UK
- MOAC DTC, University of Warwick, Coventry, UK
| | - N J Michael
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Department of Physiology, Monash University, Clayton, VIC, Australia
| | - A C Munder
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Department of Physiology, Monash University, Clayton, VIC, Australia
| | - J T Pryor
- Warwick Medical School, University of Warwick, Coventry, UK
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Department of Physiology, Monash University, Clayton, VIC, Australia
| | - L P Renaud
- Ottawa Hospital Research Institute, Ottawa Civic Hospital, Ottawa, ON, Canada
| | - D Spanswick
- NeuroSolutions Ltd, Coventry, UK
- Warwick Medical School, University of Warwick, Coventry, UK
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Department of Physiology, Monash University, Clayton, VIC, Australia
- Neuroscience Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
145
|
Bergman RN, Iyer MS. Indirect Regulation of Endogenous Glucose Production by Insulin: The Single Gateway Hypothesis Revisited. Diabetes 2017. [PMID: 28637826 DOI: 10.2337/db16-1320] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
On the basis of studies that investigated the intraportal versus systemic insulin infusion and transendothelial transport of insulin, we proposed the "single gateway hypothesis," which supposes an indirect regulation of hepatic glucose production by insulin; the rate-limiting transport of insulin across the adipose tissue capillaries is responsible for the slow suppression of free fatty acids (FFAs), which in turn is responsible for delayed suppression of hepatic endogenous glucose production (EGP) during insulin infusion. Preventing the fall in plasma FFAs during insulin infusion either by administering intralipids or by inhibiting adipose tissue lipolysis led to failure in EGP suppression, thus supporting our hypothesis. More recently, mice lacking hepatic Foxo1 in addition to Akt1 and Akt2 (L-AktFoxo1TKO), all required for insulin signaling, surprisingly showed normal glycemia. Inhibiting the fall of plasma FFAs in these mice prevented the suppression of EGP during a clamp, reaffirming that the site of insulin action to control EGP is extrahepatic. Measuring whole-body turnover rates of glucose and FFAs in L-AktFoxo1TKO mice also confirmed that hepatic EGP was regulated by insulin-mediated control of FFAs. The knockout mouse model in combination with sophisticated molecular techniques confirmed our physiological findings and the single gateway hypothesis.
Collapse
Affiliation(s)
- Richard N Bergman
- Cedars-Sinai Diabetes and Obesity Research Institute, Los Angeles, CA
| | - Malini S Iyer
- Cedars-Sinai Diabetes and Obesity Research Institute, Los Angeles, CA
| |
Collapse
|
146
|
Deem JD, Muta K, Scarlett JM, Morton GJ, Schwartz MW. How Should We Think About the Role of the Brain in Glucose Homeostasis and Diabetes? Diabetes 2017; 66:1758-1765. [PMID: 28603139 PMCID: PMC5482090 DOI: 10.2337/dbi16-0067] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 02/25/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Jennifer D Deem
- Department of Medicine, University of Washington Diabetes Institute, University of Washington, Seattle, WA
| | - Kenjiro Muta
- Department of Medicine, University of Washington Diabetes Institute, University of Washington, Seattle, WA
| | - Jarrad M Scarlett
- Department of Medicine, University of Washington Diabetes Institute, University of Washington, Seattle, WA
| | - Gregory J Morton
- Department of Medicine, University of Washington Diabetes Institute, University of Washington, Seattle, WA
| | - Michael W Schwartz
- Department of Medicine, University of Washington Diabetes Institute, University of Washington, Seattle, WA
| |
Collapse
|
147
|
Heni M, Wagner R, Kullmann S, Gancheva S, Roden M, Peter A, Stefan N, Preissl H, Häring HU, Fritsche A. Hypothalamic and Striatal Insulin Action Suppresses Endogenous Glucose Production and May Stimulate Glucose Uptake During Hyperinsulinemia in Lean but Not in Overweight Men. Diabetes 2017; 66:1797-1806. [PMID: 28174292 DOI: 10.2337/db16-1380] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/31/2017] [Indexed: 01/12/2023]
Abstract
Intranasal spray application facilitates insulin delivery to the human brain. Although brain insulin modulates peripheral metabolism, the mechanisms involved remain elusive. Twenty-one men underwent two hyperinsulinemic-euglycemic clamps with d-[6,6-2H2]glucose infusion to measure endogenous glucose production and glucose disappearance. On two separate days, participants received intranasal insulin or placebo. Insulin spillover into circulation after intranasal insulin application was mimicked by an intravenous insulin bolus on placebo day. On a different day, brain insulin sensitivity was assessed by functional MRI. Glucose infusion rates (GIRs) had to be increased more after nasal insulin than after placebo to maintain euglycemia in lean but not in overweight people. The increase in GIRs was associated with regional brain insulin action in hypothalamus and striatum. Suppression of endogenous glucose production by circulating insulin was more pronounced after administration of nasal insulin than after placebo. Furthermore, glucose uptake into tissue tended to be higher after nasal insulin application. No such effects were detected in overweight participants. By increasing insulin-mediated suppression of endogenous glucose production and stimulating peripheral glucose uptake, brain insulin may improve glucose metabolism during systemic hyperinsulinemia. Obese people appear to lack these mechanisms. Therefore, brain insulin resistance in obesity may have unfavorable consequences for whole-body glucose homeostasis.
Collapse
Affiliation(s)
- Martin Heni
- Division of Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry, Department of Internal Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Robert Wagner
- Division of Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry, Department of Internal Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Stephanie Kullmann
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Sofiya Gancheva
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Michael Roden
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Andreas Peter
- Division of Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry, Department of Internal Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Norbert Stefan
- Division of Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry, Department of Internal Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Hubert Preissl
- Division of Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry, Department of Internal Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Pharmaceutical Sciences, Department of Pharmacy and Biochemistry, Eberhard Karls University Tübingen, Tübingen, Germany
- Interfaculty Centre for Pharmacogenomics and Pharma Research, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Hans-Ulrich Häring
- Division of Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry, Department of Internal Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Interfaculty Centre for Pharmacogenomics and Pharma Research, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Division of Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry, Department of Internal Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| |
Collapse
|
148
|
Kistenmacher A, Manneck S, Wardzinski EK, Martens JC, Gohla G, Melchert UH, Jauch-Chara K, Oltmanns KM. Persistent blood glucose reduction upon repeated transcranial electric stimulation in men. Brain Stimul 2017; 10:780-786. [DOI: 10.1016/j.brs.2017.03.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 03/20/2017] [Accepted: 03/22/2017] [Indexed: 02/06/2023] Open
|
149
|
Ruud J, Steculorum SM, Brüning JC. Neuronal control of peripheral insulin sensitivity and glucose metabolism. Nat Commun 2017; 8:15259. [PMID: 28469281 PMCID: PMC5418592 DOI: 10.1038/ncomms15259] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 03/14/2017] [Indexed: 12/19/2022] Open
Abstract
The central nervous system (CNS) has an important role in the regulation of peripheral insulin sensitivity and glucose homeostasis. Research in this dynamically developing field has progressed rapidly due to techniques allowing targeted transgenesis and neurocircuitry mapping, which have defined the primary responsive neurons, associated molecular mechanisms and downstream neurocircuitries and processes involved. Here we review the brain regions, neurons and molecular mechanisms by which the CNS controls peripheral glucose metabolism, particularly via regulation of liver, brown adipose tissue and pancreatic function, and highlight the potential implications of these regulatory pathways in type 2 diabetes and obesity. The brain controls peripheral glucose metabolism, for example by modulating hepatic gluconeogenesis or by regulating glucose uptake into brown adipose tissue. Here, the authors review the brain regions, neurons and molecular mechanisms involved in these processes, and discuss their relevance to disease.
Collapse
Affiliation(s)
- Johan Ruud
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| | - Sophie M. Steculorum
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| | - Jens C. Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
- National Center for Diabetes Research (DZD), Ingolstädter Land Strasse 1, 85764 Neuherberg, Germany
| |
Collapse
|
150
|
Sugiyama M, Banno R, Mizoguchi A, Tominaga T, Tsunekawa T, Onoue T, Hagiwara D, Ito Y, Morishita Y, Iwama S, Goto M, Suga H, Arima H. PTP1B deficiency improves hypothalamic insulin sensitivity resulting in the attenuation of AgRP mRNA expression under high-fat diet conditions. Biochem Biophys Res Commun 2017; 488:116-121. [PMID: 28479249 DOI: 10.1016/j.bbrc.2017.05.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 05/03/2017] [Indexed: 11/19/2022]
Abstract
Hypothalamic insulin receptor signaling regulates energy balance and glucose homeostasis via agouti-related protein (AgRP). While protein tyrosine phosphatase 1B (PTP1B) is classically known to be a negative regulator of peripheral insulin signaling by dephosphorylating both insulin receptor β (IRβ) and insulin receptor substrate, the role of PTP1B in hypothalamic insulin signaling remains to be fully elucidated. In the present study, we investigated the role of PTP1B in hypothalamic insulin signaling using PTP1B deficient (KO) mice in vivo and ex vivo. For the in vivo study, hypothalamic insulin resistance induced by a high-fat diet (HFD) improved in KO mice compared to wild-type (WT) mice. Hypothalamic AgRP mRNA expression levels were also significantly decreased in KO mice independent of body weight changes. In an ex vivo study using hypothalamic organotypic cultures, insulin treatment significantly increased the phosphorylation of both IRβ and Akt in the hypothalamus of KO mice compared to WT mice, and also significantly decreased AgRP mRNA expression levels in KO mice. While incubation with inhibitors of phosphatidylinositol-3 kinase (PI3K) had no effect on basal levels of Akt phosphorylation, these suppressed insulin induction of Akt phosphorylation to almost basal levels in WT and KO mice. The inhibition of the PI3K-Akt pathway blocked the downregulation of AgRP mRNA expression in KO mice treated with insulin. These data suggest that PTP1B acts on the hypothalamic insulin signaling via the PI3K-Akt pathway. Together, our results suggest a deficiency of PTP1B improves hypothalamic insulin sensitivity resulting in the attenuation of AgRP mRNA expression under HFD conditions.
Collapse
Affiliation(s)
- Mariko Sugiyama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Ryoichi Banno
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.
| | - Akira Mizoguchi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Takashi Tominaga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Taku Tsunekawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Takeshi Onoue
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Daisuke Hagiwara
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yoshihiro Ito
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yoshiaki Morishita
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Shintaro Iwama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; Research Center of Health, Physical Fitness and Sports, Nagoya University, Nagoya 464-8601, Japan
| | - Motomitsu Goto
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| |
Collapse
|