1
|
Li X, Yang Y, Bai X, Wang X, Tan H, Chen Y, Zhu Y, Liu Q, Wu MN, Li Y. A brain-derived insulin signal encodes protein satiety for nutrient-specific feeding inhibition. Cell Rep 2024; 43:114282. [PMID: 38795342 PMCID: PMC11220824 DOI: 10.1016/j.celrep.2024.114282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 04/08/2024] [Accepted: 05/10/2024] [Indexed: 05/27/2024] Open
Abstract
The suppressive effect of insulin on food intake has been documented for decades. However, whether insulin signals can encode a certain type of nutrients to regulate nutrient-specific feeding behavior remains elusive. Here, we show that in female Drosophila, a pair of dopaminergic neurons, tritocerebrum 1-dopaminergic neurons (T1-DANs), are directly activated by a protein-intake-induced insulin signal from insulin-producing cells (IPCs). Intriguingly, opto-activating IPCs elicits feeding inhibition for both protein and sugar, while silencing T1-DANs blocks this inhibition only for protein food. Elevating insulin signaling in T1-DANs or opto-activating these neurons is sufficient to mimic protein satiety. Furthermore, this signal is conveyed to local neurons of the protocerebral bridge (PB-LNs) and specifically suppresses protein intake. Therefore, our findings reveal that a brain-derived insulin signal encodes protein satiety and suppresses feeding behavior in a nutrient-specific manner, shedding light on the functional specificity of brain insulin signals in regulating behaviors.
Collapse
Affiliation(s)
- Xiaoyu Li
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Science, Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Yang
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Science, Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaobing Bai
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Science, Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Sino-Danish Center for Education and Research, Beijing 100190, China
| | - Xiaotong Wang
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Science, Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Houqi Tan
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Science, Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanbo Chen
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Science, Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China
| | - Yan Zhu
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Science, Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Sino-Danish Center for Education and Research, Beijing 100190, China
| | - Qili Liu
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Mark N Wu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yan Li
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Science, Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Sino-Danish Center for Education and Research, Beijing 100190, China.
| |
Collapse
|
2
|
Todorovic S, Simeunovic V, Prvulovic M, Dakic T, Jevdjovic T, Sokanovic S, Kanazir S, Mladenovic A. Dietary restriction alters insulin signaling pathway in the brain. Biofactors 2024; 50:450-466. [PMID: 37975613 DOI: 10.1002/biof.2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/07/2023] [Indexed: 11/19/2023]
Abstract
Insulin is known to be a key hormone in the regulation of peripheral glucose homeostasis, but beyond that, its effects on the brain are now undisputed. Impairments in insulin signaling in the brain, including changes in insulin levels, are thought to contribute significantly to declines in cognitive performance, especially during aging. As one of the most widely studied experimental interventions, dietary restriction (DR) is considered to delay the neurodegenerative processes associated with aging. Recently, however, data began to suggest that the onset and duration of a restrictive diet play a critical role in the putative beneficial outcome. Because the effects of DR on insulin signaling in the brain have been poorly studied, we decided to examine the effects of DR that differed in onset and duration: long-term DR (LTDR), medium-term DR (MTDR), and short-term DR (STDR) on the expression of proteins involved in insulin signaling in the hippocampus of 18- and 24-month-old male Wistar rats. We found that DR-induced changes in insulin levels in the brain may be independent of what happens in the periphery after restricted feeding. Significantly changed insulin content in the hippocampus, together with altered insulin signaling were found under the influence of DR, but the outcome was highly dependent on the onset and duration of DR.
Collapse
Affiliation(s)
- Smilja Todorovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Valentina Simeunovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milica Prvulovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tamara Dakic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry "Ivan Djaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Tanja Jevdjovic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry "Ivan Djaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Srdjan Sokanovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Selma Kanazir
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Mladenovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
3
|
Dakic TB, Markelic MB, Ruzicic AA, Jevdjovic TV, Lakic IV, Djordjevic JD, Vujovic PZ. Hypothalamic insulin expression remains unaltered after short-term fasting in female rats. Endocrine 2022; 78:476-483. [PMID: 36301508 DOI: 10.1007/s12020-022-03235-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/15/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE Our previous study showed that 6-h fasting increased insulin expression in the hypothalamus of male rats. We, therefore, wanted to examine if this phenomenon occurs in female rats and whether it depended on the estrus cycle phase. METHODS Female rats in proestrus or diestrus were either exposed to 6-h fasting or had ad libitum access to food. The serum, cerebrospinal fluid, and hypothalamic insulin levels were determined using radioimmunoassay. The hypothalamic insulin mRNA expression was measured by RT-qPCR, while the hypothalamic insulin distribution was assessed immunohistochemically. RESULTS Albeit the short-term fasting lowered circulating insulin, both hypothalamic insulin mRNA expression and hypothalamic insulin content remained unaltered. As for the hypothalamic insulin distribution, strong insulin immunopositivity was noted primarily in ependymal cells lining the upper part of the third ventricle and some neurons mainly located within the periventricular nucleus. The pattern of insulin distribution was similar between the controls and the females exposed to fasting regardless of the estrous cycle phase. CONCLUSION The findings of this study indicate that the control of insulin expression in the hypothalamus differs from that in the pancreatic beta cells during short-term fasting. Furthermore, they also imply that the regulation of insulin expression in the female hypothalamus is different from males but independent of the estrus cycle phase.
Collapse
Affiliation(s)
- Tamara B Dakic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry Ivan Djaja, University of Belgrade-Faculty of Biology, Studentski trg 16, 11000, Belgrade, Serbia.
| | - Milica B Markelic
- Department of Cell and Tissue Biology, Institute for Zoology, University of Belgrade-Faculty of Biology, Studentski trg 16, 11000, Belgrade, Serbia
| | - Aleksandra A Ruzicic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry Ivan Djaja, University of Belgrade-Faculty of Biology, Studentski trg 16, 11000, Belgrade, Serbia
| | - Tanja V Jevdjovic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry Ivan Djaja, University of Belgrade-Faculty of Biology, Studentski trg 16, 11000, Belgrade, Serbia
| | - Iva V Lakic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry Ivan Djaja, University of Belgrade-Faculty of Biology, Studentski trg 16, 11000, Belgrade, Serbia
| | - Jelena D Djordjevic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry Ivan Djaja, University of Belgrade-Faculty of Biology, Studentski trg 16, 11000, Belgrade, Serbia
| | - Predrag Z Vujovic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry Ivan Djaja, University of Belgrade-Faculty of Biology, Studentski trg 16, 11000, Belgrade, Serbia
| |
Collapse
|
4
|
Finnell JE, Ferrario CR. Intra-NAc insulin reduces the motivation for food and food intake without altering cue-triggered food-seeking. Physiol Behav 2022; 254:113892. [PMID: 35753434 PMCID: PMC10583176 DOI: 10.1016/j.physbeh.2022.113892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 11/24/2022]
Abstract
Insulin receptors are expressed throughout the adult brain, and insulin from the periphery reaches the central nervous system. In humans and rodents, actions of insulin in the brain decrease food intake. Furthermore, insulin receptor activation alters dopamine and glutamate transmission within mesolimbic regions that influence food-seeking and feeding including the nucleus accumbens (NAc). Here we determined how intra-NAc insulin affects conditioned approach (a measure of cue-triggered food-seeking), free food intake, and the motivation to obtain food in hungry rats using Pavlovian and instrumental approaches. Intra-NAc insulin did not affect conditioned approach but did reduce home cage chow intake immediately following conditioned approach testing. Consistent with reduced chow intake, intra-NAc insulin also reduced the motivation to work for flavored food pellets (assessed by a progressive ratio procedure). This effect was partially reversed by insulin receptor blockade and was not driven by insulin-induced sickness or malaise. Taken together, these data show that insulin within the NAc does not alter behavioral responses to a food cue, but instead reduces the motivation to work for and consume food in hungry animals. These data are discussed in light of insulin's role in the regulation of feeding, and its dysregulation by obesity.
Collapse
Affiliation(s)
- Julie E Finnell
- Department of Pharmacology, University of Michigan, United States
| | - Carrie R Ferrario
- Department of Pharmacology, University of Michigan, United States; Psychology Department (Biopsychology), University of Michigan, Ann Arbor MI 48109, United States.
| |
Collapse
|
5
|
Agrawal M, Agrawal AK. Pathophysiological Association Between Diabetes Mellitus and Alzheimer's Disease. Cureus 2022; 14:e29120. [PMID: 36258952 PMCID: PMC9559718 DOI: 10.7759/cureus.29120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/13/2022] [Indexed: 12/05/2022] Open
Abstract
Worldwide elderly people are being affected by diabetes mellitus (DM) and dementia. The risk for the development of dementia is higher in people with DM. DM causes a marked cognitive reduction and increases the risk of dementia, most commonly vascular dementia and Alzheimer's disease. People affected by DM and dementia seem to be at higher risk for intense hypoglycemia. Hypoglycemia, the complication of DM treatment, is believed as an independent risk factor for dementia in people with DM. Both Alzheimer's disease and DM are linked with decreased insulin secretion, reduced uptake of glucose, raised oxidative stress, angiopathy, activation of the apoptotic pathway, aging, abnormal peroxidation of lipids, increased production of advanced glycation end products and tau phosphorylation, brain atrophy, and decreased fat metabolism. In this paper, we will review the association between Alzheimer's disease and DM. In addition, we will discuss the agents that enhance the risk for dementia in elderly people with DM and how to prevent the development of cognitive dysfunction in DM.
Collapse
|
6
|
Meier DT, Rachid L, Wiedemann SJ, Traub S, Trimigliozzi K, Stawiski M, Sauteur L, Winter DV, Le Foll C, Brégère C, Guzman R, Odermatt A, Böni-Schnetzler M, Donath MY. Prohormone convertase 1/3 deficiency causes obesity due to impaired proinsulin processing. Nat Commun 2022; 13:4761. [PMID: 35963866 PMCID: PMC9376086 DOI: 10.1038/s41467-022-32509-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 08/03/2022] [Indexed: 12/12/2022] Open
Abstract
Defective insulin processing is associated with obesity and diabetes. Prohormone convertase 1/3 (PC1/3) is an endopeptidase required for the processing of neurotransmitters and hormones. PC1/3 deficiency and genome-wide association studies relate PC1/3 with early onset obesity. Here, we find that deletion of PC1/3 in obesity-related neuronal cells expressing proopiomelanocortin mildly and transiently change body weight and fail to produce a phenotype when targeted to Agouti-related peptide- or nestin-expressing tissues. In contrast, pancreatic β cell-specific PC1/3 ablation induces hyperphagia with consecutive obesity despite uncontrolled diabetes with glucosuria. Obesity develops not due to impaired pro-islet amyloid polypeptide processing but due to impaired insulin maturation. Proinsulin crosses the blood-brain-barrier but does not induce central satiety. Accordingly, insulin therapy prevents hyperphagia. Further, islet PC1/3 expression levels negatively correlate with body mass index in humans. In this work, we show that impaired PC1/3-mediated proinsulin processing, as observed in human prediabetes, promotes hyperphagic obesity. Defective insulin secretion is observed early in the development of diabetes. Here the authors report that β cell-specific deficiency of the insulin prohormone convertase 1/3 (PC1/3) leads not only to hyperglycemia, but also to hyperphagic obesity in mice.
Collapse
Affiliation(s)
- Daniel T Meier
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland. .,Department of Biomedicine, University of Basel, Basel, Switzerland.
| | - Leila Rachid
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Sophia J Wiedemann
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Shuyang Traub
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Kelly Trimigliozzi
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marc Stawiski
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Loïc Sauteur
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Denise V Winter
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Christelle Le Foll
- Institute of Veterinary Physiology, University of Zurich, 8057, Zurich, Switzerland
| | - Catherine Brégère
- Department of Biomedicine, University of Basel, Basel, Switzerland.,Department of Neurosurgery, University of Basel, Basel, Switzerland
| | - Raphael Guzman
- Department of Biomedicine, University of Basel, Basel, Switzerland.,Department of Neurosurgery, University of Basel, Basel, Switzerland
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Marianne Böni-Schnetzler
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marc Y Donath
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
7
|
Competing paradigms of obesity pathogenesis: energy balance versus carbohydrate-insulin models. Eur J Clin Nutr 2022; 76:1209-1221. [PMID: 35896818 PMCID: PMC9436778 DOI: 10.1038/s41430-022-01179-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 02/07/2023]
Abstract
The obesity pandemic continues unabated despite a persistent public health campaign to decrease energy intake (“eat less”) and increase energy expenditure (“move more”). One explanation for this failure is that the current approach, based on the notion of energy balance, has not been adequately embraced by the public. Another possibility is that this approach rests on an erroneous paradigm. A new formulation of the energy balance model (EBM), like prior versions, considers overeating (energy intake > expenditure) the primary cause of obesity, incorporating an emphasis on “complex endocrine, metabolic, and nervous system signals” that control food intake below conscious level. This model attributes rising obesity prevalence to inexpensive, convenient, energy-dense, “ultra-processed” foods high in fat and sugar. An alternative view, the carbohydrate-insulin model (CIM), proposes that hormonal responses to highly processed carbohydrates shift energy partitioning toward deposition in adipose tissue, leaving fewer calories available for the body’s metabolic needs. Thus, increasing adiposity causes overeating to compensate for the sequestered calories. Here, we highlight robust contrasts in how the EBM and CIM view obesity pathophysiology and consider deficiencies in the EBM that impede paradigm testing and refinement. Rectifying these deficiencies should assume priority, as a constructive paradigm clash is needed to resolve long-standing scientific controversies and inform the design of new models to guide prevention and treatment. Nevertheless, public health action need not await resolution of this debate, as both models target processed carbohydrates as major drivers of obesity.
Collapse
|
8
|
Pradhan LK, Sahoo PK, Chauhan S, Das SK. Recent Advances Towards Diagnosis and Therapeutic Fingerprinting for Alzheimer's Disease. J Mol Neurosci 2022; 72:1143-1165. [PMID: 35553375 DOI: 10.1007/s12031-022-02009-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/02/2022] [Indexed: 12/12/2022]
Abstract
Since the report of "a peculiar severe disease process of the cerebral cortex" by Alois Alzheimer in 1906, it was considered to be a rare condition characterized by loss of cognition, memory impairment, and pathological markers such as senile plaques or neurofibrillary tangles (NFTs). Later on, the report was published in the textbook "Psychiatrie" and the disease was named as Alzheimer's disease (AD) and was known to be the consequences of aging; however, owing to its complex etiology, there is no cure for the progressive neurodegenerative disorder. Our current understanding of the mechanisms involved in the pathogenesis of AD is still at the mechanistic level. The treatment strategies applied currently only alleviate the symptoms and co-morbidities. For instance, the available treatments such as the usage of acetylcholinesterase inhibitors and N-methyl D-aspartate antagonists have minimal impact on the disease progression and target the later aspects of the disease. The recent advancements in the last two decades have made us more clearly understand the pathophysiology of the disease which has led to the development of novel therapeutic strategies. This review gives a brief idea about the various facets of AD pathophysiology and its management through modern investigational therapies to give a new direction for development of targeted therapeutic measures.
Collapse
Affiliation(s)
- Lilesh Kumar Pradhan
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Kalinga Nagar, Bhubaneswar-751003, India
| | - Pradyumna Kumar Sahoo
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Kalinga Nagar, Bhubaneswar-751003, India
| | - Santosh Chauhan
- Autophagy Laboratory, Infectious Disease Biology Division, Institute of Life Sciences, Bhubaneswar-751023, India.
| | - Saroj Kumar Das
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Kalinga Nagar, Bhubaneswar-751003, India.
| |
Collapse
|
9
|
Milstein JL, Ferris HA. The brain as an insulin-sensitive metabolic organ. Mol Metab 2021; 52:101234. [PMID: 33845179 PMCID: PMC8513144 DOI: 10.1016/j.molmet.2021.101234] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/26/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The brain was once thought of as an insulin-insensitive organ. We now know that the insulin receptor is present throughout the brain and serves important functions in whole-body metabolism and brain function. Brain insulin signaling is involved not only in brain homeostatic processes but also neuropathological processes such as cognitive decline and Alzheimer's disease. SCOPE OF REVIEW In this review, we provide an overview of insulin signaling within the brain and the metabolic impact of brain insulin resistance and discuss Alzheimer's disease, one of the neurologic diseases most closely associated with brain insulin resistance. MAJOR CONCLUSIONS While brain insulin signaling plays only a small role in central nervous system glucose regulation, it has a significant impact on the brain's metabolic health. Normal insulin signaling is important for mitochondrial functioning and normal food intake. Brain insulin resistance contributes to obesity and may also play an important role in neurodegeneration.
Collapse
Affiliation(s)
- Joshua L Milstein
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA; Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Heather A Ferris
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA; Department of Neuroscience, University of Virginia, Charlottesville, VA, USA; Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
10
|
Rojas M, Chávez-Castillo M, Pirela D, Parra H, Nava M, Chacín M, Angarita L, Añez R, Salazar J, Ortiz R, Durán Agüero S, Gravini-Donado M, Bermúdez V, Díaz-Camargo E. Metabolic Syndrome: Is It Time to Add the Central Nervous System? Nutrients 2021; 13:nu13072254. [PMID: 34208833 PMCID: PMC8308252 DOI: 10.3390/nu13072254] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/05/2021] [Accepted: 06/09/2021] [Indexed: 12/28/2022] Open
Abstract
Metabolic syndrome (MS) is a set of cardio-metabolic risk factors that includes central obesity, hyperglycemia, hypertension, and dyslipidemias. The syndrome affects 25% of adults worldwide. The definition of MS has evolved over the last 80 years, with various classification systems and criteria, whose limitations and benefits are currently the subject of some controversy. Likewise, hypotheses regarding the etiology of MS add more confusion from clinical and epidemiological points of view. The leading suggestion for the pathophysiology of MS is insulin resistance (IR). IR can affect multiple tissues and organs, from the classic “triumvirate” (myocyte, adipocyte, and hepatocyte) to possible effects on organs considered more recently, such as the central nervous system (CNS). Mild cognitive impairment (MCI) and Alzheimer’s disease (AD) may be clinical expressions of CNS involvement. However, the association between MCI and MS is not understood. The bidirectional relationship that seems to exist between these factors raises the questions of which phenomenon occurs first and whether MCI can be a precursor of MS. This review explores shared pathophysiological mechanisms between MCI and MS and establishes a hypothesis of a possible MCI role in the development of IR and the appearance of MS.
Collapse
Affiliation(s)
- Milagros Rojas
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.R.); (D.P.); (H.P.); (M.N.); (J.S.)
| | | | - Daniela Pirela
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.R.); (D.P.); (H.P.); (M.N.); (J.S.)
| | - Heliana Parra
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.R.); (D.P.); (H.P.); (M.N.); (J.S.)
| | - Manuel Nava
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.R.); (D.P.); (H.P.); (M.N.); (J.S.)
| | - Maricarmen Chacín
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla 08002, Colombia;
| | - Lissé Angarita
- Escuela de Nutrición y Dietética, Facultad de Medicina, Universidad Andrés Bello, Sede Concepción 4260000, Chile;
| | - Roberto Añez
- Departamento de Endocrinología y Nutrición, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain;
| | - Juan Salazar
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.R.); (D.P.); (H.P.); (M.N.); (J.S.)
| | - Rina Ortiz
- Posgrado, Carrera de Medicina, Universidad Católica de Cuenca, Cantón de Cuenca 010101, Ecuador;
| | - Samuel Durán Agüero
- Facultad de Ciencias Para el Cuidado de la Salud, Universidad San Sebastián, Los Leones 8420524, Chile;
| | - Marbel Gravini-Donado
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Barranquilla 080002, Colombia;
| | - Valmore Bermúdez
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Cúcuta 540006, Colombia;
| | - Edgar Díaz-Camargo
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Cúcuta 540006, Colombia;
- Correspondence:
| |
Collapse
|
11
|
Zhang AM, Wellberg EA, Kopp JL, Johnson JD. Hyperinsulinemia in Obesity, Inflammation, and Cancer. Diabetes Metab J 2021; 45:285-311. [PMID: 33775061 PMCID: PMC8164941 DOI: 10.4093/dmj.2020.0250] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022] Open
Abstract
The relative insufficiency of insulin secretion and/or insulin action causes diabetes. However, obesity and type 2 diabetes mellitus can be associated with an absolute increase in circulating insulin, a state known as hyperinsulinemia. Studies are beginning to elucidate the cause-effect relationships between hyperinsulinemia and numerous consequences of metabolic dysfunctions. Here, we review recent evidence demonstrating that hyperinsulinemia may play a role in inflammation, aging and development of cancers. In this review, we will focus on the consequences and mechanisms of excess insulin production and action, placing recent findings that have challenged dogma in the context of the existing body of literature. Where relevant, we elaborate on the role of specific signal transduction components in the actions of insulin and consequences of chronic hyperinsulinemia. By discussing the involvement of hyperinsulinemia in various metabolic and other chronic diseases, we may identify more effective therapeutics or lifestyle interventions for preventing or treating obesity, diabetes and cancer. We also seek to identify pertinent questions that are ripe for future investigation.
Collapse
Affiliation(s)
- Anni M.Y. Zhang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Elizabeth A. Wellberg
- Department of Pathology, University of Oklahoma Health Sciences Center, Stephenson Cancer Center, Harold Hamm Diabetes Center, Oklahoma City, OK, USA
| | - Janel L. Kopp
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - James D. Johnson
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
12
|
Abstract
The intranasal (IN) route enables the delivery of insulin to the central nervous system in the relative absence of systemic uptake and related peripheral side effects. Intranasally administered insulin is assumed to travel along olfactory and adjacent pathways and has been shown to rapidly accumulate in cerebrospinal fluid, indicating efficient transport to the brain. Two decades of studies in healthy humans and patients have demonstrated that IN insulin exerts functional effects on metabolism, such as reductions in food intake and body weight and improvements of glucose homeostasis, as well as cognition, ie, enhancements of memory performance both in healthy individuals and patients with mild cognitive impairment or Alzheimer's disease; these studies moreover indicate a favourable safety profile of the acute and repeated use of IN insulin. Emerging findings suggest that IN insulin also modulates neuroendocrine activity, sleep-related mechanisms, sensory perception and mood. Some, but not all studies point to sex differences in the response to IN insulin that need to be further investigated along with the impact of age. "Brain insulin resistance" is an evolving concept that posits impairments in central nervous insulin signalling as a pathophysiological factor in metabolic and cognitive disorders such as obesity, type 2 diabetes and Alzheimer's disease, and, notably, a target of interventions that rely on IN insulin. Still, the negative outcomes of longer-term IN insulin trials in individuals with obesity or Alzheimer's disease highlight the need for conceptual as well as methodological advances to translate the promising results of proof-of-concept experiments and pilot clinical trials into the successful clinical application of IN insulin.
Collapse
Affiliation(s)
- Manfred Hallschmid
- Institute of Medical Psychology and Behavioural Neurobiology, University of Tübingen, Tübingen, Germany
- German Centre for Diabetes Research (DZD), Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
| |
Collapse
|
13
|
Fujikawa T. Central regulation of glucose metabolism in an insulin-dependent and -independent manner. J Neuroendocrinol 2021; 33:e12941. [PMID: 33599044 DOI: 10.1111/jne.12941] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/17/2022]
Abstract
The central nervous system (CNS) contributes significantly to glucose homeostasis. The available evidence indicates that insulin directly acts on the CNS, in particular the hypothalamus, to regulate hepatic glucose production, thereby controlling whole-body glucose metabolism. Additionally, insulin also acts on the brain to regulate food intake and fat metabolism, which may indirectly regulate glucose metabolism. Studies conducted over the last decade have found that the CNS can regulate glucose metabolism in an insulin-independent manner. Enhancement of central leptin signalling reverses hyperglycaemia in insulin-deficient rodents. Here, I review the mechanisms by which central insulin and leptin actions regulate glucose metabolism. Although clinical studies have shown that insulin treatment is currently indispensable for managing diabetes, unravelling the neuronal mechanisms underlying the central regulation of glucose metabolism will pave the way for the design of novel therapeutic drugs for diabetes.
Collapse
Affiliation(s)
- Teppei Fujikawa
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
14
|
Wiedemann SJ, Rachid L, Illigens B, Böni-Schnetzler M, Donath MY. Evidence for cephalic phase insulin release in humans: A systematic review and meta-analysis. Appetite 2020; 155:104792. [DOI: 10.1016/j.appet.2020.104792] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/24/2020] [Accepted: 06/24/2020] [Indexed: 02/02/2023]
|
15
|
Hu S, Wang L, Togo J, Yang D, Xu Y, Wu Y, Douglas A, Speakman JR. The carbohydrate-insulin model does not explain the impact of varying dietary macronutrients on the body weight and adiposity of mice. Mol Metab 2019; 32:27-43. [PMID: 32029228 PMCID: PMC6938849 DOI: 10.1016/j.molmet.2019.11.010] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 11/08/2019] [Indexed: 12/24/2022] Open
Abstract
Objectives The carbohydrate-insulin model (CIM) predicts that increases in fasting and post-prandial insulin in response to dietary carbohydrates stimulate energy intake and lower energy expenditures, leading to positive energy balance and weight gain. The objective of the present study was to directly test the CIM's predictions using C57BL/6 mice. Methods Diets were designed by altering dietary carbohydrates with either fixed protein or fat content and were fed to C57BL/6 mice acutely or chronically for 12 weeks. The body weight, body composition, food intake, and energy expenditures of the mice were measured. Their fasting and post-prandial glucose and insulin levels were also measured. RNA-seq was performed on RNA from the hypothalamus and subcutaneous white adipose tissue. Pathway analysis was conducted using IPA. Results Only the post-prandial insulin and fasting glucose levels followed the CIM's predictions. The lipolysis and leptin signaling pathways in the sWAT were inhibited in relation to the elevated fasting insulin, supporting the CIM's predicted impact of high insulin. However, because higher fasting insulin was unrelated to carbohydrate intake, the overall pattern did not support the model. Moreover, the hypothalamic hunger pathways were inhibited in relation to the increased fasting insulin, and the energy intake was not increased. The browning pathway in the sWAT was inhibited at higher insulin levels, but the daily energy expenditure was not altered. Conclusions Two of the predictions were partially supported (and hence also partially not supported) and the other three predictions were not supported. We conclude that the CIM does not explain the impact of dietary macronutrients on adiposity in mice. Higher fasting insulin related to inhibited lipolysis and leptin pathways in sWAT, supporting CIM. Higher fasting insulin related to inhibited hypothalamic hunger pathway, contrasting CIM. Fasting insulin decreased with higher dietary carbohydrate, overall contrasting CIM. Higher dietary carbohydrate did not lead to greater EI/adiposity, or lowered EE.
Collapse
Affiliation(s)
- Sumei Hu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Lu Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China; University of Chinese Academy of Sciences, Shijingshan District, Beijing, 100049, PR China; Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, AB24 2TZ, Scotland, UK
| | - Jacques Togo
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China; University of Chinese Academy of Sciences, Shijingshan District, Beijing, 100049, PR China
| | - Dengbao Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Yanchao Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Yingga Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China; University of Chinese Academy of Sciences, Shijingshan District, Beijing, 100049, PR China; Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, AB24 2TZ, Scotland, UK
| | - Alex Douglas
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, AB24 2TZ, Scotland, UK
| | - John R Speakman
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China; Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, AB24 2TZ, Scotland, UK; CAS Center for Excellence in Animal Evolution and Genetics (CCEAEG), Kunming, PR China.
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Nakhate KT, Subhedar NK, Kokare DM. Involvement of neuropeptide CART in the central effects of insulin on feeding and body weight. Pharmacol Biochem Behav 2019; 181:101-109. [PMID: 31054945 DOI: 10.1016/j.pbb.2019.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 04/23/2019] [Accepted: 05/01/2019] [Indexed: 12/15/2022]
Abstract
While insulin secreted from pancreas plays a pivotal role in the control of glucose homeostasis, it also interacts with hypothalamic sites and negatively influences the energy balance. The present study was undertaken to reveal the functional interaction between cocaine- and amphetamine-regulated transcript (CART), a well-known anorexic peptide, and insulin within the framework of hypothalamus in the regulation of feeding behavior and body weight. Insulin was administered daily by intracerebroventricular (icv) route, alone or in combination with CART (icv) for a period of seven days. Immediately thereafter, preweighed food was offered to the animals at the commencement of the dark phase. The food intake and body weight were measured daily just prior to next injection. Furthermore, brains of insulin-treated rats were processed for the immunohistochemical analysis of CART-containing elements in the hypothalamus. Treatment with insulin (6 mU, icv) for a period of 7 days caused a significant decrease in food intake and body weight as compared to control. Concomitant administration of CART (0.5 μg, icv) potentiated insulin-induced anorexia and weight loss. Insulin administration resulted in a significant increase in CART immunoreactivity in the hypothalamic arcuate, paraventricular, dorsomedial and ventromedial nuclei. We suggest that increased CART contents in the hypothalamus may be causally linked with anorexia and weight loss induced by insulin.
Collapse
Affiliation(s)
- Kartik T Nakhate
- Rungta College of Pharmaceutical Sciences and Research, Rungta Educational Campus, Kohka-Kurud Road, Bhilai 490 024, Chhattisgarh, India
| | - Nishikant K Subhedar
- Indian Institute of Science Education and Research (IISER), Dr. Homi Bhabha Road, Near NCL, Pune 411 021, Maharashtra, India
| | - Dadasaheb M Kokare
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, Maharashtra, India.
| |
Collapse
|
19
|
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
|
20
|
Molecular Connection Between Diabetes and Dementia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1128:103-131. [DOI: 10.1007/978-981-13-3540-2_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
21
|
Castellani LN, Wilkin J, Abela AR, Benarroch L, Ahasan Z, Teo C, Wilson V, Kowalchuk C, Giacca A, Remington GJ, Hahn MK. Effects of acute olanzapine exposure on central insulin-mediated regulation of whole body fuel selection and feeding. Psychoneuroendocrinology 2018; 98:127-130. [PMID: 30142549 DOI: 10.1016/j.psyneuen.2018.07.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 07/13/2018] [Accepted: 07/29/2018] [Indexed: 01/01/2023]
Abstract
The use of antipsychotics is associated with severe disruptions in whole body glucose and lipid metabolism which may in part occur through the central nervous system and impaired insulin action at the brain. Here we investigated whether olanzapine treatment might also affect the ability of central insulin treatment to regulate food intake and fuel preference in the light and dark cycle. Male Sprague-Dawley rats were treated with olanzapine (or vehicle solution; 3 mg/kg, subcutaneous) and a simultaneous acute intracerebral ventricular (ICV) infusion of insulin (or vehicle; 3 μL at 10mU; ICV) at the beginning of the 12-h light and dark cycles. Olanzapine treatment reduced RER in the dark and light phases (most consistently in the 4-hours post-treatment), while ICV insulin reduced average RER predominantly in the dark phase, but also at the end of the light cycle. The RER lowering effect of ICV-insulin during the light cycle was absent in the group co-administered olanzapine. The reduction in RER during the dark phase was mirrored by decreased food intake with ICV insulin, but not olanzapine treated rats. The reduction in food intake by ICV-insulin was abolished in rats co-administered olanzapine suggesting rapid induction of central insulin resistance. A combination of ICV-insulin and olanzapine similarly reduced RER in the dark phase, independent of changes in food intake. Olanzapine treatment, alone or in combination with ICV-insulin, significantly reduced VCO2 at regular intervals in the dark phase (specifically 3 h post-treatment), while VO2 was not significantly altered by either treatment. Finally, heat production was increased by olanzapine treatment in the light phase, though this effect was not consistent. The findings confirm that acute olanzapine treatment directly reduces RER and suggest that treatment with this drug may also override central insulin-mediated reductions in food intake at the hypothalamus (while still independently favoring fatty acid oxidation). Acute central insulin similarly reduces RER, but in contrast to olanzapine, this may represent a physiologically appropriate response to reduction in food intake.
Collapse
Affiliation(s)
- Laura N Castellani
- Centre for Addiction and Mental Health, 250 College St, Toronto, ON, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, ON, M5T 1R8, Canada
| | - Jennifer Wilkin
- Centre for Addiction and Mental Health, 250 College St, Toronto, ON, M5T 1R8, Canada.
| | - Andrew R Abela
- Centre for Addiction and Mental Health, 250 College St, Toronto, ON, M5T 1R8, Canada
| | - Louise Benarroch
- Centre for Addiction and Mental Health, 250 College St, Toronto, ON, M5T 1R8, Canada
| | - Zohra Ahasan
- Centre for Addiction and Mental Health, 250 College St, Toronto, ON, M5T 1R8, Canada.
| | - Celine Teo
- Centre for Addiction and Mental Health, 250 College St, Toronto, ON, M5T 1R8, Canada
| | - Virginia Wilson
- Centre for Addiction and Mental Health, 250 College St, Toronto, ON, M5T 1R8, Canada.
| | - Chantel Kowalchuk
- Centre for Addiction and Mental Health, 250 College St, Toronto, ON, M5T 1R8, Canada; Institute of Medical Sciences, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8T, Canada.
| | - Adria Giacca
- Department of Physiology, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada; Institute of Medical Sciences, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8T, Canada; Banting and Best Diabetes Centre, Eaton Building, Room 12E248, 200 Elizabeth St, Toronto, ON M5G 2C4, Canada.
| | - Gary J Remington
- Centre for Addiction and Mental Health, 250 College St, Toronto, ON, M5T 1R8, Canada; Institute of Medical Sciences, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8T, Canada; Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, ON, M5T 1R8, Canada
| | - Margaret K Hahn
- Centre for Addiction and Mental Health, 250 College St, Toronto, ON, M5T 1R8, Canada; Institute of Medical Sciences, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8T, Canada; Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, ON, M5T 1R8, Canada; Banting and Best Diabetes Centre, Eaton Building, Room 12E248, 200 Elizabeth St, Toronto, ON M5G 2C4, Canada.
| |
Collapse
|
22
|
Onaolapo A, Onaolapo O. Food additives, food and the concept of ‘food addiction’: Is stimulation of the brain reward circuit by food sufficient to trigger addiction? PATHOPHYSIOLOGY 2018; 25:263-276. [DOI: 10.1016/j.pathophys.2018.04.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 02/26/2018] [Accepted: 04/07/2018] [Indexed: 02/08/2023] Open
|
23
|
Mueller PL, Pritchett CE, Wiechman TN, Zharikov A, Hajnal A. Antidepressant-like effects of insulin and IGF-1 are mediated by IGF-1 receptors in the brain. Brain Res Bull 2018; 143:27-35. [DOI: 10.1016/j.brainresbull.2018.09.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/14/2018] [Accepted: 09/25/2018] [Indexed: 12/11/2022]
|
24
|
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
|
25
|
Nguyen TTL, Chan LC, Borreginne K, Kale RP, Hu C, Tye SJ. A review of brain insulin signaling in mood disorders: From biomarker to clinical target. Neurosci Biobehav Rev 2018; 92:7-15. [PMID: 29758232 DOI: 10.1016/j.neubiorev.2018.05.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 04/08/2018] [Accepted: 05/08/2018] [Indexed: 12/16/2022]
Abstract
Patients with mood disorders are at increased risk for metabolic dysfunction. Co-occurrence of the two conditions is typically associated with a more severe disease course and poorer treatment outcomes. The specific pathophysiological mechanisms underlying this bidirectional relationship between mood and metabolic dysfunction remains poorly understood. However, it is likely that impairment of metabolic processes within the brain play a critical role. The insulin signaling pathway mediates metabolic homeostasis and is important in the regulation of neurotrophic and synaptic plasticity processes, including those involved in neurodegenerative diseases like Alzheimer's. Thus, insulin signaling in the brain may serve to link metabolic function and mood. Central insulin signaling is mediated through locally secreted insulin and widespread insulin receptor expression. Here we review the preclinical and clinical data addressing the relationships between central insulin signaling, cellular metabolism, neurotrophic processes, and mood regulation, including key points of mechanistic overlap. These relationships have important implications for developing biomarker-based diagnostics and precision medicine approaches to treat severe mood disorders.
Collapse
Affiliation(s)
- Thanh Thanh L Nguyen
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, United States; Department of Biology and Psychology, Green Mountain College, 1 Brennan Cir, Poultney, VT, 05764, United States
| | - Lily C Chan
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, United States
| | - Kristin Borreginne
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, United States
| | - Rajas P Kale
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, United States; School of Engineering, Deakin University, Waurn Ponds, VIC, 3216, Australia
| | - Chunling Hu
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, United States
| | - Susannah J Tye
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, United States; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, United States; Department of Psychiatry, University of Minnesota, 3 Morrill Hall, 100 Church Street SE, Minneapolis, MN, 55454, United States; School of Psychology, Deakin University, Burwood, VIC, 3125, Australia; Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.
| |
Collapse
|
26
|
Benedict C, Grillo CA. Insulin Resistance as a Therapeutic Target in the Treatment of Alzheimer's Disease: A State-of-the-Art Review. Front Neurosci 2018; 12:215. [PMID: 29743868 PMCID: PMC5932355 DOI: 10.3389/fnins.2018.00215] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/19/2018] [Indexed: 01/10/2023] Open
Abstract
Research in animals and humans has shown that type 2 diabetes and its prodromal state, insulin resistance, promote major pathological hallmarks of Alzheimer's disease (AD), such as the formation of amyloid plaques and neurofibrillary tangles (NFT). Worrisomely, dysregulated amyloid beta (Aβ) metabolism has also been shown to promote central nervous system insulin resistance; although the role of tau metabolism remains controversial. Collectively, as proposed in this review, these findings suggest the existence of a mechanistic interplay between AD pathogenesis and disrupted insulin signaling. They also provide strong support for the hypothesis that pharmacologically restoring brain insulin signaling could represent a promising strategy to curb the development and progression of AD. In this context, great hopes have been attached to the use of intranasal insulin. This drug delivery method increases cerebrospinal fluid concentrations of insulin in the absence of peripheral side effects, such as hypoglycemia. With this in mind, the present review will also summarize current knowledge on the efficacy of intranasal insulin to mitigate major pathological symptoms of AD, i.e., cognitive impairment and deregulation of Aβ and tau metabolism.
Collapse
Affiliation(s)
| | - Claudia A Grillo
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina-School of Medicine, Columbia, SC, United States
| |
Collapse
|
27
|
Vasselli JR, Pi-Sunyer FX, Wall DG, John CS, Chapman CD, Currie PJ. Central effects of insulin detemir on feeding, body weight, and metabolism in rats. Am J Physiol Endocrinol Metab 2017; 313:E613-E621. [PMID: 28720583 PMCID: PMC5792141 DOI: 10.1152/ajpendo.00111.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 07/11/2017] [Accepted: 07/11/2017] [Indexed: 12/20/2022]
Abstract
Insulin detemir (DET) is a basal insulin analog that, in contrast to other long-acting forms of insulin, has significant weight-gain-sparing effects in diabetic patients. We hypothesized that this effect of DET may be due to its enhanced catabolic action in the central nervous system. We investigated the long-term effects of single third ventricular (3V) microinjections of equimolar doses of DET and regular insulin in normal male rats on feeding, body weight, energy expenditure (EE), and respiratory quotient (RQ). Also, in acute testing, we assessed the ability of lower doses of DET to alter feeding, EE, and RQ when microinjected directly into the paraventricular nucleus (PVN). The anabolic peptide ghrelin served as a positive control in acute testing. 3V administration of both DET (0.5-2.0 mU) and regular insulin (2.0-8.0 mU) significantly reduced feeding and body weight over 48 and 120 h, respectively, with DET yielding greater inhibitory effects. DET also stimulated greater elevations of EE and reductions of RQ over 72 and 48 h postinjection, respectively. In acute (4 h) testing, microinjections of DET (0.5 mU) into the PVN reduced feeding, increased EE, and reduced RQ, while ghrelin (100 pmol) had the opposite effects. When administered sequentially into the PVN, DET (0.25 and 0.5 mU) reversed ghrelin-induced feeding, EE, and RQ effects. These data support the notion that the weight-sparing effect of DET is at least in part based on its central catabolic action and that enhanced EE and reduced RQ may participate in this effect.
Collapse
Affiliation(s)
- Joseph R Vasselli
- Obesity Nutrition Research Center, Department of Medicine, Columbia University, New York, New York; and
| | - F Xavier Pi-Sunyer
- Obesity Nutrition Research Center, Department of Medicine, Columbia University, New York, New York; and
| | - Daniel G Wall
- Department of Psychology, Reed College, Portland, Oregon
| | | | | | - Paul J Currie
- Department of Psychology, Reed College, Portland, Oregon
| |
Collapse
|
28
|
LPS-Induced Low-Grade Inflammation Increases Hypothalamic JNK Expression and Causes Central Insulin Resistance Irrespective of Body Weight Changes. Int J Mol Sci 2017; 18:ijms18071431. [PMID: 28677618 PMCID: PMC5535922 DOI: 10.3390/ijms18071431] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/22/2017] [Accepted: 06/27/2017] [Indexed: 01/26/2023] Open
Abstract
Metabolic endotoxemia contributes to low-grade inflammation in obesity, which causes insulin resistance due to the activation of intracellular proinflammatory pathways, such as the c-Jun N-terminal Kinase (JNK) cascade in the hypothalamus and other tissues. However, it remains unclear whether the proinflammatory process precedes insulin resistance or it appears because of the development of obesity. Hypothalamic low-grade inflammation was induced by prolonged lipopolysaccharide (LPS) exposure to investigate if central insulin resistance is induced by an inflammatory stimulus regardless of obesity. Male Wistar rats were treated with single (1 LPS) or repeated injections (6 LPS) of LPS (100 μg/kg, IP) to evaluate the phosphorylation of the insulin receptor substrate-1 (IRS1), Protein kinase B (AKT), and JNK in the hypothalamus. Single LPS increased the expression of pIRS1, pAKT, and pJNK, whereas the repeated LPS treatment failed to recruit pIRS1 and pAKT. The 6 LPS treated rats showed increased total JNK and pJNK. The 6 LPS rats became unresponsive to the hypophagic effect induced by central insulin administration (12 μM/5 μL, ICV). Prolonged exposure to LPS (24 h) impaired the insulin-induced AKT phosphorylation and the translocation of the transcription factor forkhead box protein O1 (FoxO1) from the nucleus to the cytoplasm of the cultured hypothalamic GT1-7 cells. Central administration of the JNK inhibitor (20 μM/5 μL, ICV) restored the ability of insulin to phosphorylate IRS1 and AKT in 6 LPS rats. The present data suggest that an increased JNK activity in the hypothalamus underlies the development of insulin resistance during prolonged exposure to endotoxins. Our study reveals that weight gain is not mandatory for the development of hypothalamic insulin resistance and the blockade of proinflammatory pathways could be useful for restoring the insulin signaling during prolonged low-grade inflammation as seen in obesity.
Collapse
|
29
|
Hlavica M, Delparente A, Good A, Good N, Plattner PS, Seyedsadr MS, Schwab ME, Figlewicz DP, Ineichen BV. Intrathecal insulin-like growth factor 1 but not insulin enhances myelin repair in young and aged rats. Neurosci Lett 2017; 648:41-46. [PMID: 28363754 DOI: 10.1016/j.neulet.2017.03.047] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/16/2017] [Accepted: 03/28/2017] [Indexed: 12/29/2022]
Abstract
One main pathological hallmark of multiple sclerosis (MS) is demyelination. Novel therapies which enhance myelin repair are urgently needed. Insulin and insulin-like growth factor 1 (IGF-1) have strong functional relationships. Here, we addressed the potential capacity of IGF-1 and insulin to enhance remyelination in an animal demyelination model in vivo. We found that chronic intrathecal infusion of IGF-1 enhanced remyelination after lysolecithin-induced demyelination in the spinal cord of young and aged rats. Aged rats showed a weaker innate remyelination capacity and are therefore a good model for progressive MS which is defined by chronic demyelination. In contrast to IGF-1, Insulin had no effect on remyelination in either age group. Our findings highlight the potential use of IGF-1 as remyelinating therapy for MS, particularly the progressive stage in which chronic demyelination is the hallmark.
Collapse
Affiliation(s)
- Martin Hlavica
- Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, ETH Zurich, 8057 Zurich, Switzerland; Cantonal Hospital St.Gallen, Department of Neurosurgery, Switzerland
| | - Aro Delparente
- Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, ETH Zurich, 8057 Zurich, Switzerland
| | - Andrin Good
- Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, ETH Zurich, 8057 Zurich, Switzerland
| | - Nicolas Good
- Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, ETH Zurich, 8057 Zurich, Switzerland
| | - Patricia S Plattner
- Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, ETH Zurich, 8057 Zurich, Switzerland
| | - Maryam S Seyedsadr
- Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, ETH Zurich, 8057 Zurich, Switzerland
| | - Martin E Schwab
- Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, ETH Zurich, 8057 Zurich, Switzerland
| | - Dianne P Figlewicz
- VA Puget Sound Health Care System, University of Washington, Seattle, WA, USA
| | - Benjamin V Ineichen
- Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, ETH Zurich, 8057 Zurich, Switzerland; University Hospital Zurich, Department of Neurology, 8091 Zurich, Switzerland.
| |
Collapse
|
30
|
Belfort-DeAguiar R, Seo D, Naik S, Hwang J, Lacadie C, Schmidt C, Constable RT, Sinha R, Sherwin R. Food image-induced brain activation is not diminished by insulin infusion. Int J Obes (Lond) 2016; 40:1679-1686. [PMID: 27569684 PMCID: PMC5101182 DOI: 10.1038/ijo.2016.152] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 07/26/2016] [Accepted: 08/07/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND/OBJECTIVES The obesity epidemic appears to be driven in large part by our modern environment inundated by food cues, which may influence our desire to eat. Although insulin decreases food intake in both animals and humans, the effect of insulin on motivation for food in the presence of food cues is not known. Therefore, the aim of this study was to evaluate the effect of an intravenous insulin infusion on the brain response to visual food cues, hunger and food craving in non-obese human subjects. SUBJECTS/METHODS Thirty-four right-handed healthy non-obese subjects (19F/15M, age: 29±8 years.; BMI: 23.1±2.1 kg m-2) were divided in two groups matched by age and BMI; the insulin group (18 subjects) underwent a hyperinsulinemic-euglycemic-clamp, and the control group (16 subjects) received an intravenous saline infusion, while viewing high and low-calorie food and non-food pictures during a functional MRI scan. Motivation for food was determined via analog scales for hunger, wanting and liking ratings. RESULTS Food images induced brain responses in the hypothalamus, striatum, amygdala, insula, ventromedial prefrontal cortex (PFC), dorsolateral PFC and occipital lobe (whole brain correction, P<0.05). Wanting (P<0.001) and liking (P<0.001) ratings were significantly higher for the food than the non-food images, but not different between insulin and saline infusion groups. Hunger ratings increased throughout the MRI scan and correlated with preference for high-calorie food pictures (r=0.70; P<0.001). However, neither brain activity nor food cravings were affected by hyperinsulinemia or hormonal status (leptin and ghrelin levels) (P=NS). CONCLUSIONS Our data demonstrate that visual food cues induce a strong response in motivation/reward and cognitive-executive control brain regions in non-obese subjects, but that these responses are not diminished by hyperinsulinemia per se. These findings suggest that our modern food cue saturated environment may be sufficient to overpower homeostatic hormonal signals, and thus contribute to the current obesity epidemic.
Collapse
Affiliation(s)
| | - Dongju Seo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Sarita Naik
- Section of Endocrinology, Yale University School of Medicine, New Haven, CT, USA
- University College London Hospitals NHS, London, UK
| | - Janice Hwang
- Section of Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| | - Cheryl Lacadie
- Department of Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Christian Schmidt
- Section of Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| | - R. Todd Constable
- Department of Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Rajita Sinha
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Robert Sherwin
- Section of Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
31
|
Stanley M, Macauley SL, Holtzman DM. Changes in insulin and insulin signaling in Alzheimer's disease: cause or consequence? J Exp Med 2016; 213:1375-85. [PMID: 27432942 PMCID: PMC4986537 DOI: 10.1084/jem.20160493] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/20/2016] [Indexed: 12/30/2022] Open
Abstract
Stanley and colleagues explore whether changes in insulin levels and insulin
signaling are a cause or consequence of AD. Individuals with type 2 diabetes have an increased risk for developing
Alzheimer’s disease (AD), although the causal relationship remains poorly
understood. Alterations in insulin signaling (IS) are reported in the AD brain.
Moreover, oligomers/fibrils of amyloid-β (Aβ) can lead to neuronal
insulin resistance and intranasal insulin is being explored as a potential therapy
for AD. Conversely, elevated insulin levels (ins) are found in AD patients and high
insulin has been reported to increase Aβ levels and tau phosphorylation, which
could exacerbate AD pathology. Herein, we explore whether changes in ins and IS are a
cause or consequence of AD.
Collapse
Affiliation(s)
- Molly Stanley
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| | - Shannon L Macauley
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
32
|
Habash T, Eskay R, Kuenzel W, Castonguay T. Interactions of Glucocorticoids, NPY and Hypothalamic Serotonin. Nutr Neurosci 2016; 3:183-92. [DOI: 10.1080/1028415x.2000.11747315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
33
|
Khanh DV, Choi YH, Moh SH, Kinyua AW, Kim KW. Leptin and insulin signaling in dopaminergic neurons: relationship between energy balance and reward system. Front Psychol 2014; 5:846. [PMID: 25147530 PMCID: PMC4124796 DOI: 10.3389/fpsyg.2014.00846] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 07/16/2014] [Indexed: 01/28/2023] Open
Abstract
The central actions of leptin and insulin are essential for the regulation of energy and glucose homeostasis. In addition to the crucial effects on the hypothalamus, emerging evidence suggests that the leptin and insulin signaling can act on other brain regions to mediate the reward value of nutrients. Recent studies have indicated the midbrain dopaminergic neurons as a potential site for leptin' and insulin's actions on mediating the feeding behaviors and therefore affecting the energy balance. Although molecular details about the integrative roles of leptin and insulin in this subset of neurons remain to be investigated, substantial body of evidence by far imply that the signaling pathways regulated by leptin and insulin may play an essential role in the regulation of energy balance through the control of food-associated reward. This review therefore describes the convergence of energy regulation and reward system, particularly focusing on leptin and insulin signaling in the midbrain dopaminergic neurons.
Collapse
Affiliation(s)
- Doan V. Khanh
- Departments of Pharmacology and Global Biomedical Science, Wonju College of Medicine, Yonsei UniversityWonju, South Korea
- Institute of Lifestyle Medicine and Nuclear Receptor Research Consortium, Wonju College of Medicine, Yonsei UniversityWonju, South Korea
| | - Yun-Hee Choi
- Institute of Lifestyle Medicine and Nuclear Receptor Research Consortium, Wonju College of Medicine, Yonsei UniversityWonju, South Korea
- Antiaging Research Institute of BIO-FD&C Co. Ltd.Incheon, South Korea
| | - Sang Hyun Moh
- Antiaging Research Institute of BIO-FD&C Co. Ltd.Incheon, South Korea
| | - Ann W. Kinyua
- Departments of Pharmacology and Global Biomedical Science, Wonju College of Medicine, Yonsei UniversityWonju, South Korea
- Institute of Lifestyle Medicine and Nuclear Receptor Research Consortium, Wonju College of Medicine, Yonsei UniversityWonju, South Korea
| | - Ki Woo Kim
- Departments of Pharmacology and Global Biomedical Science, Wonju College of Medicine, Yonsei UniversityWonju, South Korea
- Institute of Lifestyle Medicine and Nuclear Receptor Research Consortium, Wonju College of Medicine, Yonsei UniversityWonju, South Korea
| |
Collapse
|
34
|
Woods SC. Metabolic signals and food intake. Forty years of progress. Appetite 2013; 71:440-4. [DOI: 10.1016/j.appet.2012.08.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 07/11/2012] [Accepted: 08/15/2012] [Indexed: 11/16/2022]
|
35
|
Pomytkin IA. H2O2 Signalling Pathway: A Possible Bridge between Insulin Receptor and Mitochondria. Curr Neuropharmacol 2013; 10:311-20. [PMID: 23730255 PMCID: PMC3520041 DOI: 10.2174/157015912804143559] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Revised: 06/07/2012] [Accepted: 06/24/2012] [Indexed: 01/20/2023] Open
Abstract
This review is focused on the mechanistic aspects of the insulin-induced H2O2 signalling pathway in neurons and the molecules affecting it, which act as risk factors for developing central insulin resistance. Insulin-induced H2O2 promotes insulin receptor activation and the mitochondria act as the insulin-sensitive H2O2 source, providing a direct molecular link between mitochondrial dysfunction and irregular insulin receptor activation. In this view, the accumulation of dysfunctional mitochondria during chronological ageing and Alzheimer's disease (AD) is a risk factor that may contribute to the development of dysfunctional cerebral insulin receptor signalling and insulin resistance. Due to the high significance of insulin-induced H2O2 for insulin receptor activation, oxidative stress-induced upregulation of antioxidant enzymes, e.g., in AD brains, may represent another risk factor contributing to the development of insulin resistance. As insulin-induced H2O2 signalling requires fully functional mitochondria, pharmacological strategies based on activating mitochondria biogenesis in the brain are central to the treatment of diseases associated with dysfunctional insulin receptor signalling in this organ.
Collapse
|
36
|
No effect of adjunctive, repeated dose intranasal insulin treatment on body metabolism in patients with schizophrenia. Schizophr Res 2013; 146:40-5. [PMID: 23434504 PMCID: PMC3622829 DOI: 10.1016/j.schres.2013.01.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Revised: 01/22/2013] [Accepted: 01/28/2013] [Indexed: 11/22/2022]
Abstract
OBJECTIVE This study examined the effect of adjunctive intranasal insulin therapy on body metabolism in patients with schizophrenia. METHOD Each subject had a DSM-IV diagnosis of schizophrenia or schizoaffective disorder and had been on stable dose of antipsychotic agent for at least one month. In an 8-week randomized, double-blind, placebo-controlled study, subjects received either intranasal insulin (40 IU 4 times per day) or placebo. The whole body dual-energy X-ray absorptiometry (DXA) was used to assess body composition. Lipid particles were assessed using nuclear magnetic resonance (NMR) spectroscopy. All assessments were conducted at baseline, and repeated at week 8. RESULTS A total number of 39 subjects completed the study (18 in the insulin group, 21 in the placebo group). There were no significant differences between the two groups in week 8 changes for body weight, body mass index, waist circumference, as well as various measures of lipid particles (p's>0.100). The DXA assessment showed no significant differences between the two groups in week 8 changes for fat mass, lean mass or total mass (p's>0.100). CONCLUSION In the present study, adjunctive therapy of intranasal insulin did not seem to improve body metabolism in patients with schizophrenia. The implications for future studies were discussed.
Collapse
|
37
|
Begg DP, Mul JD, Liu M, Reedy BM, D'Alessio DA, Seeley RJ, Woods SC. Reversal of diet-induced obesity increases insulin transport into cerebrospinal fluid and restores sensitivity to the anorexic action of central insulin in male rats. Endocrinology 2013; 154:1047-54. [PMID: 23337529 PMCID: PMC3578991 DOI: 10.1210/en.2012-1929] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Diet-induced obesity (DIO) reduces the ability of centrally administered insulin to reduce feeding behavior and also reduces the transport of insulin from the periphery to the central nervous system (CNS). The current study was designed to determine whether reversal of high-fat DIO restores the anorexic efficacy of central insulin and whether this is accompanied by restoration of the compromised insulin transport. Adult male Long-Evans rats were initially maintained on either a low-fat chow diet (LFD) or a high-fat diet (HFD). After 22 weeks, half of the animals on the HFD were changed to the LFD, whereas the other half continued on the HFD for an additional 8 weeks, such that there were 3 groups: 1) a LFD control group (Con; n = 18), 2) a HFD-fed, DIO group (n = 17), and 3) a HFD to LFD, DIO-reversal group (DIO-rev; n = 18). The DIO reversal resulted in a significant reduction of body weight and epididymal fat weight relative to the DIO group. Acute central insulin administration (8 mU) reduced food intake and caused weight loss in Con and DIO-rev but not DIO rats. Fasting cerebrospinal fluid insulin was higher in DIO than Con animals. However, after a peripheral bolus injection of insulin, cerebrospinal fluid insulin increased in Con and DIO-rev rats but not in the DIO group. These data provide support for previous reports that DIO inhibits both the central effects of insulin and insulin's transport to the CNS. Importantly, DIO-rev restored sensitivity to the effects of central insulin on food intake and insulin transport into the CNS.
Collapse
Affiliation(s)
- Denovan P Begg
- Departments of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45237, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Scherer T, Lehnert H, Hallschmid M. Brain insulin and leptin signaling in metabolic control: from animal research to clinical application. Endocrinol Metab Clin North Am 2013; 42:109-25. [PMID: 23391243 DOI: 10.1016/j.ecl.2012.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Besides the well-characterized effects of brain insulin and leptin in regulating food intake, insulin and leptin signaling to the central nervous system modulates a variety of metabolic processes, such as glucose and lipid homeostasis, as well as energy expenditure. This review summarizes the current literature on the contribution of central nervous insulin and leptin action to metabolic control in animals and humans. Potential therapeutic options based on the direct delivery of these peptides to the brain by, for example, intranasal administration, are discussed.
Collapse
Affiliation(s)
- Thomas Scherer
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna 1090, Austria.
| | | | | |
Collapse
|
39
|
Begg DP, Woods SC. Interactions between the central nervous system and pancreatic islet secretions: a historical perspective. ADVANCES IN PHYSIOLOGY EDUCATION 2013; 37:53-60. [PMID: 23471249 PMCID: PMC3776474 DOI: 10.1152/advan.00167.2012] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 01/14/2013] [Indexed: 05/10/2023]
Abstract
The endocrine pancreas is richly innervated with sympathetic and parasympathetic projections from the brain. In the mid-20th century, it was established that α-adrenergic activation inhibits, whereas cholinergic stimulation promotes, insulin secretion; this demonstrated the importance of the sympathetic and parasympathetic systems in pancreatic endocrine function. It was later established that insulin injected peripherally could act within the brain, leading to the discovery of insulin and insulin receptors within the brain and the receptor-mediated transport of insulin into the central nervous system from endothelial cells. The insulin receptor within the central nervous system is widely distributed, reflecting insulin's diverse range of actions, including acting as an adiposity signal to reduce food intake and increase energy expenditure, regulation of systemic glucose responses, altering sympathetic activity, and involvement in cognitive function. As observed with central insulin administration, the pancreatic hormones glucagon, somatostatin, pancreatic polypeptide, and amylin can each also reduce food intake. Pancreatic and also gut hormones are released cephalically, in what is an important mechanism to prepare the body for a meal and prevent excessive postprandial hyperglycemia.
Collapse
Affiliation(s)
- Denovan P Begg
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45237, USA
| | | |
Collapse
|
40
|
Vogt MC, Brüning JC. CNS insulin signaling in the control of energy homeostasis and glucose metabolism - from embryo to old age. Trends Endocrinol Metab 2013; 24:76-84. [PMID: 23265947 DOI: 10.1016/j.tem.2012.11.004] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 11/07/2012] [Accepted: 11/13/2012] [Indexed: 10/27/2022]
Abstract
Central nervous system (CNS) insulin signaling regulates energy and glucose homeostasis by acting on hypothalamic neurocircuits and higher brain circuits such as the dopaminergic system. However, overnutrition, obesity, and type 2 diabetes mellitus (T2DM) induce insulin resistance selectively in different regions of the brain, thereby impairing energy homeostasis and augmenting disease progression. Moreover, fetal hyperinsulinemia in response to maternal overnutrition, obesity, and diabetes disrupts hypothalamic neurocircuit development and predisposes to metabolic disorders later in life. In light of the current obesity and diabetes epidemic, we review the molecular basis of insulin action and resistance in the CNS, mechanisms which are causal to the development of these metabolic disorders, both in the neonate and in the adult.
Collapse
Affiliation(s)
- Merly C Vogt
- Max Planck Institute for Neurological Research, D-50931 Cologne, Germany
| | | |
Collapse
|
41
|
Aimé P, Hegoburu C, Jaillard T, Degletagne C, Garcia S, Messaoudi B, Thevenet M, Lorsignol A, Duchamp C, Mouly AM, Julliard AK. A physiological increase of insulin in the olfactory bulb decreases detection of a learned aversive odor and abolishes food odor-induced sniffing behavior in rats. PLoS One 2012; 7:e51227. [PMID: 23251461 PMCID: PMC3522659 DOI: 10.1371/journal.pone.0051227] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 10/30/2012] [Indexed: 11/19/2022] Open
Abstract
Insulin is involved in multiple regulatory mechanisms, including body weight and food intake, and plays a critical role in metabolic disorders such as obesity and diabetes. An increasing body of evidence indicates that insulin is also involved in the modulation of olfactory function. The olfactory bulb (OB) contains the highest level of insulin and insulin receptors (IRs) in the brain. However, a role for insulin in odor detection and sniffing behavior remains to be elucidated. Using a behavioral paradigm based on conditioned olfactory aversion (COA) to isoamyl-acetate odor, we demonstrated that an intracerebroventricular (ICV) injection of 14 mU insulin acutely decreased olfactory detection of fasted rats to the level observed in satiated animals. In addition, whereas fasted animals demonstrated an increase in respiratory frequency upon food odor detection, this effect was absent in fasted animals receiving a 14 mU insulin ICV injection as well as in satiated animals. In parallel, we showed that the OB and plasma insulin levels were increased in satiated rats compared to fasted rats, and that a 14 mU insulin ICV injection elevated the OB insulin level of fasted rats to that of satiated rats. We further quantified insulin receptors (IRs) distribution and showed that IRs are preferentially expressed in the caudal and lateral parts of the main OB, with the highest labeling found in the mitral cells, the main OB projection neurons. Together, these data suggest that insulin acts on the OB network to modulate olfactory processing and demonstrate that olfactory function is under the control of signals involved in energy homeostasis regulation and feeding behaviors.
Collapse
Affiliation(s)
- Pascaline Aimé
- Centre de Recherche en Neurosciences de Lyon (CRNL), Team Olfaction: From Coding to Memory, CNRS UMR 5292 - INSERM U1028- Université Lyon1 - Université de Lyon, Lyon, France
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| | - Chloé Hegoburu
- Centre de Recherche en Neurosciences de Lyon (CRNL), Team Olfaction: From Coding to Memory, CNRS UMR 5292 - INSERM U1028- Université Lyon1 - Université de Lyon, Lyon, France
| | - Tristan Jaillard
- Métabolisme Plasticité Mitochondrie, CNRS UMR 5241 - Université Paul Sabatier, Toulouse, France
- STROMALab, CNRS UMR 5273 - EFS - INSERM U1031- Université Paul Sabatier, Toulouse, France
| | - Cyril Degletagne
- Laboratoire d'Ecologie des Hydrosystèmes Naturels et Anthropisés (LEHNA), CNRS UMR 5023 - Université Lyon 1 – Université de Lyon, Villeurbanne, France
| | - Samuel Garcia
- Centre de Recherche en Neurosciences de Lyon (CRNL), Team Olfaction: From Coding to Memory, CNRS UMR 5292 - INSERM U1028- Université Lyon1 - Université de Lyon, Lyon, France
| | - Belkacem Messaoudi
- Centre de Recherche en Neurosciences de Lyon (CRNL), Team Olfaction: From Coding to Memory, CNRS UMR 5292 - INSERM U1028- Université Lyon1 - Université de Lyon, Lyon, France
| | - Marc Thevenet
- Centre de Recherche en Neurosciences de Lyon (CRNL), Team Olfaction: From Coding to Memory, CNRS UMR 5292 - INSERM U1028- Université Lyon1 - Université de Lyon, Lyon, France
| | - Anne Lorsignol
- Métabolisme Plasticité Mitochondrie, CNRS UMR 5241 - Université Paul Sabatier, Toulouse, France
- STROMALab, CNRS UMR 5273 - EFS - INSERM U1031- Université Paul Sabatier, Toulouse, France
| | - Claude Duchamp
- Laboratoire d'Ecologie des Hydrosystèmes Naturels et Anthropisés (LEHNA), CNRS UMR 5023 - Université Lyon 1 – Université de Lyon, Villeurbanne, France
| | - Anne-Marie Mouly
- Centre de Recherche en Neurosciences de Lyon (CRNL), Team Olfaction: From Coding to Memory, CNRS UMR 5292 - INSERM U1028- Université Lyon1 - Université de Lyon, Lyon, France
| | - Andrée Karyn Julliard
- Centre de Recherche en Neurosciences de Lyon (CRNL), Team Olfaction: From Coding to Memory, CNRS UMR 5292 - INSERM U1028- Université Lyon1 - Université de Lyon, Lyon, France
- * E-mail:
| |
Collapse
|
42
|
Palouzier-Paulignan B, Lacroix MC, Aimé P, Baly C, Caillol M, Congar P, Julliard AK, Tucker K, Fadool DA. Olfaction under metabolic influences. Chem Senses 2012; 37:769-97. [PMID: 22832483 PMCID: PMC3529618 DOI: 10.1093/chemse/bjs059] [Citation(s) in RCA: 230] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Recently published work and emerging research efforts have suggested that the olfactory system is intimately linked with the endocrine systems that regulate or modify energy balance. Although much attention has been focused on the parallels between taste transduction and neuroendocrine controls of digestion due to the novel discovery of taste receptors and molecular components shared by the tongue and gut, the equivalent body of knowledge that has accumulated for the olfactory system, has largely been overlooked. During regular cycles of food intake or disorders of endocrine function, olfaction is modulated in response to changing levels of various molecules, such as ghrelin, orexins, neuropeptide Y, insulin, leptin, and cholecystokinin. In view of the worldwide health concern regarding the rising incidence of diabetes, obesity, and related metabolic disorders, we present a comprehensive review that addresses the current knowledge of hormonal modulation of olfactory perception and how disruption of hormonal signaling in the olfactory system can affect energy homeostasis.
Collapse
Affiliation(s)
- Brigitte Palouzier-Paulignan
- Centre de Recherche des Neurosciences de Lyon, Equipe Olfaction du Codage à la Mémoire, INSERM U 1028/CNRS 5292, Université de Lyon150 Ave. Tony Garnier, 69366, Lyon, Cedex 07,France
- Equal contribution
| | - Marie-Christine Lacroix
- INRA, UR1197 Neurobiologie de l’Olfaction et Modélisation en ImagerieF-78350, Jouy-en-JosasFrance
- IFR 144NeuroSud Paris, 91190 Gif-Sur-YvetteFrance
- Equal contribution
| | - Pascaline Aimé
- Centre de Recherche des Neurosciences de Lyon, Equipe Olfaction du Codage à la Mémoire, INSERM U 1028/CNRS 5292, Université de Lyon150 Ave. Tony Garnier, 69366, Lyon, Cedex 07,France
| | - Christine Baly
- INRA, UR1197 Neurobiologie de l’Olfaction et Modélisation en ImagerieF-78350, Jouy-en-JosasFrance
- IFR 144NeuroSud Paris, 91190 Gif-Sur-YvetteFrance
| | - Monique Caillol
- INRA, UR1197 Neurobiologie de l’Olfaction et Modélisation en ImagerieF-78350, Jouy-en-JosasFrance
- IFR 144NeuroSud Paris, 91190 Gif-Sur-YvetteFrance
| | - Patrice Congar
- INRA, UR1197 Neurobiologie de l’Olfaction et Modélisation en ImagerieF-78350, Jouy-en-JosasFrance
- IFR 144NeuroSud Paris, 91190 Gif-Sur-YvetteFrance
| | - A. Karyn Julliard
- Centre de Recherche des Neurosciences de Lyon, Equipe Olfaction du Codage à la Mémoire, INSERM U 1028/CNRS 5292, Université de Lyon150 Ave. Tony Garnier, 69366, Lyon, Cedex 07,France
| | - Kristal Tucker
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of MedicinePittsburgh, PA 15261USAand
| | - Debra Ann Fadool
- Department of Biological Science, Programs in Neuroscience and Molecular Biophysics, The Florida State UniversityTallahassee, FL 32306-4295USA
| |
Collapse
|
43
|
Banks WA, Owen JB, Erickson MA. Insulin in the brain: there and back again. Pharmacol Ther 2012; 136:82-93. [PMID: 22820012 PMCID: PMC4134675 DOI: 10.1016/j.pharmthera.2012.07.006] [Citation(s) in RCA: 403] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 07/03/2012] [Indexed: 12/12/2022]
Abstract
Insulin performs unique functions within the CNS. Produced nearly exclusively by the pancreas, insulin crosses the blood-brain barrier (BBB) using a saturable transporter, affecting feeding and cognition through CNS mechanisms largely independent of glucose utilization. Whereas peripheral insulin acts primarily as a metabolic regulatory hormone, CNS insulin has an array of effects on brain that may more closely resemble the actions of the ancestral insulin molecule. Brain endothelial cells (BECs), the cells that form the vascular BBB and contain the transporter that translocates insulin from blood to brain, are themselves regulated by insulin. The insulin transporter is altered by physiological and pathological factors including hyperglycemia and the diabetic state. The latter can lead to BBB disruption. Pericytes, pluripotent cells in intimate contact with the BECs, protect the integrity of the BBB and its ability to transport insulin. Most of insulin's known actions within the CNS are mediated through two canonical pathways, the phosphoinositide-3 kinase (PI3)/Akt and Ras/mitogen activated kinase (MAPK) cascades. Resistance to insulin action within the CNS, sometimes referred to as diabetes mellitus type III, is associated with peripheral insulin resistance, but it is possible that variable hormonal resistance syndromes exist so that resistance at one tissue bed may be independent of that at others. CNS insulin resistance is associated with Alzheimer's disease, depression, and impaired baroreceptor gain in pregnancy. These aspects of CNS insulin action and the control of its entry by the BBB are likely only a small part of the story of insulin within the brain.
Collapse
Affiliation(s)
- William A Banks
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care Center, Seattle, WA, USA.
| | | | | |
Collapse
|
44
|
Ghasemi R, Haeri A, Dargahi L, Mohamed Z, Ahmadiani A. Insulin in the brain: sources, localization and functions. Mol Neurobiol 2012; 47:145-71. [PMID: 22956272 DOI: 10.1007/s12035-012-8339-9] [Citation(s) in RCA: 203] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 08/20/2012] [Indexed: 02/07/2023]
Abstract
Historically, insulin is best known for its role in peripheral glucose homeostasis, and insulin signaling in the brain has received less attention. Insulin-independent brain glucose uptake has been the main reason for considering the brain as an insulin-insensitive organ. However, recent findings showing a high concentration of insulin in brain extracts, and expression of insulin receptors (IRs) in central nervous system tissues have gathered considerable attention over the sources, localization, and functions of insulin in the brain. This review summarizes the current status of knowledge of the peripheral and central sources of insulin in the brain, site-specific expression of IRs, and also neurophysiological functions of insulin including the regulation of food intake, weight control, reproduction, and cognition and memory formation. This review also considers the neuromodulatory and neurotrophic effects of insulin, resulting in proliferation, differentiation, and neurite outgrowth, introducing insulin as an attractive tool for neuroprotection against apoptosis, oxidative stress, beta amyloid toxicity, and brain ischemia.
Collapse
Affiliation(s)
- Rasoul Ghasemi
- Department of Physiology, Faculty of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | | | | |
Collapse
|
45
|
Electrophysiological analysis of circuits controlling energy homeostasis. Mol Neurobiol 2012; 45:258-78. [PMID: 22331510 DOI: 10.1007/s12035-012-8241-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 01/26/2012] [Indexed: 10/28/2022]
Abstract
Since the discovery of leptin and the central melanocortin circuit, electrophysiological studies have played a major role in elucidating mechanisms underlying energy homeostasis. This review highlights the contribution of findings made by electrophysiological measurements to the current understanding of hypothalamic neuronal networks involved in energy homeostasis with a specific focus on the arcuate-paraventricular nucleus circuit.
Collapse
|
46
|
Ryan KK, Woods SC, Seeley RJ. Central nervous system mechanisms linking the consumption of palatable high-fat diets to the defense of greater adiposity. Cell Metab 2012; 15:137-49. [PMID: 22244528 PMCID: PMC3278569 DOI: 10.1016/j.cmet.2011.12.013] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 09/01/2011] [Accepted: 10/03/2011] [Indexed: 12/23/2022]
Abstract
The central nervous system (CNS) plays key role in the homeostatic regulation of body weight. Satiation and adiposity signals, providing acute and chronic information about available fuel, are produced in the periphery and act in the brain to influence energy intake and expenditure, resulting in the maintenance of stable adiposity. Diet-induced obesity (DIO) does not result from a failure of these central homeostatic circuits. Rather, the threshold for defended adiposity is increased in environments providing ubiquitous access to palatable, high-fat foods, making it difficult to achieve and maintain weight loss. Consequently, mechanisms by which nutritional environments interact with central homeostatic circuits to influence the threshold for defended adiposity represent critical targets for therapeutic intervention.
Collapse
Affiliation(s)
- Karen K Ryan
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA
| | | | | |
Collapse
|
47
|
Abstract
Insulin acts throughout the body to reduce circulating energy and to increase energy storage. Within the brain, insulin produces a net catabolic effect by reducing food intake and increasing energy expenditure; this is evidenced by the hypophagia and increased brown adipose tissue sympathetic nerve activity induced by central insulin infusion. Reducing the activity of the brain insulin system via administration of insulin antibodies, receptor antisense treatment, or receptor knockdown results in hyperphagia and increased adiposity. However, despite decades of research into the role of central insulin in food intake, many questions remain to be answered, including the underlying mechanism of action.
Collapse
Affiliation(s)
- Denovan P Begg
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, OH 45237, USA
| | | |
Collapse
|
48
|
Mercer RE, Chee MJS, Colmers WF. The role of NPY in hypothalamic mediated food intake. Front Neuroendocrinol 2011; 32:398-415. [PMID: 21726573 DOI: 10.1016/j.yfrne.2011.06.001] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 05/17/2011] [Accepted: 06/13/2011] [Indexed: 12/29/2022]
Abstract
Neuropeptide Y (NPY) is a highly conserved neuropeptide with orexigenic actions in discrete hypothalamic nuclei that plays a role in regulating energy homeostasis. NPY signals via a family of high affinity receptors that mediate the widespread actions of NPY in all hypothalamic nuclei. These actions are also subject to tight, intricate regulation by numerous peripheral and central energy balance signals. The NPY system is embedded within a densely-redundant network designed to ensure stable energy homeostasis. This redundancy may underlie compensation for the loss of NPY or its receptors in germline knockouts, explaining why conventional knockouts of NPY or its receptors rarely yield a marked phenotypic change. We discuss insights into the hypothalamic role of NPY from studies of its physiological actions, responses to genetic manipulations and interactions with other energy balance signals. We conclude that numerous approaches must be employed to effectively study different aspects of NPY action.
Collapse
Affiliation(s)
- Rebecca E Mercer
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada T6G 2H7
| | | | | |
Collapse
|
49
|
He J, Votruba S, Venti C, Krakoff J. Higher incremental insulin area under the curve during oral glucose tolerance test predicts less food intake and weight gain. Int J Obes (Lond) 2011; 35:1495-501. [PMID: 21343902 DOI: 10.1038/ijo.2011.13] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVE To investigate the correlation of peripheral insulin concentrations with food intake and body weight. DESIGN Cross sectional and longitudinal clinical study: we investigated the association of peripheral insulin concentrations in response to an oral glucose tolerance test (OGTT) with subsequent measures of ad libitum food intake and body weight change. SUBJECTS Food intake analysis: Pima Indians (n=67, 63% male; body mass index (mean ± s.d.) 34.2 ± 9.4 kg m(-2)) with normal glucose regulation (NGR; fasting glucose <5.6 mmol l(-1) and 2-h glucose <7.8 mmol l(-1)) participated in a study of ad libitum food intake measured over 3 days by an automated vending machine system. Weight change analysis: Pima Indians with NGR (n=339) who also participated in a longitudinal study of risks for type 2 diabetes and had follow-up weights. RESULTS Food intake analysis: incremental area under the curve (iAUC) for insulin during the OGTT was negatively associated with mean daily ad libitum energy intake (DEI) (r=-0.26, P=0.04), calories consumed as percent weight-maintenance energy needs (%WMEN) (r=-0.38, P=0.002) and carbohydrate intake (gram per day) (r=-0.35, P=0.005). Adjustment for age and sex attenuated the association of iAUC with DEI (P=0.06) not with %WMEN and carbohydrate intake (P=0.005, P=0.008). Weight change analysis: after adjustment for age, sex, follow-up time and initial body weight, higher insulin iAUC predicted less absolute and percent weight change (β=-6.9, P=0.02; β=-0.08, P=0.008, respectively). CONCLUSIONS In healthy Pima Indians with NGR, higher plasma iAUC during an OGTT predicted lower food intake and carbohydrate consumption and less weight gain. These data indicated a role for peripheral insulin as a negative feedback signal in the regulation of energy intake and body weight.
Collapse
Affiliation(s)
- J He
- Department of Health and Human Services, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ 85016, USA.
| | | | | | | |
Collapse
|
50
|
Scherer T, O'Hare J, Diggs-Andrews K, Schweiger M, Cheng B, Lindtner C, Zielinski E, Vempati P, Su K, Dighe S, Milsom T, Puchowicz M, Scheja L, Zechner R, Fisher SJ, Previs SF, Buettner C. Brain insulin controls adipose tissue lipolysis and lipogenesis. Cell Metab 2011; 13:183-94. [PMID: 21284985 PMCID: PMC3061443 DOI: 10.1016/j.cmet.2011.01.008] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 09/14/2010] [Accepted: 12/06/2010] [Indexed: 01/14/2023]
Abstract
White adipose tissue (WAT) dysfunction plays a key role in the pathogenesis of type 2 diabetes (DM2). Unrestrained WAT lipolysis results in increased fatty acid release, leading to insulin resistance and lipotoxicity, while impaired de novo lipogenesis in WAT decreases the synthesis of insulin-sensitizing fatty acid species like palmitoleate. Here, we show that insulin infused into the mediobasal hypothalamus (MBH) of Sprague-Dawley rats increases WAT lipogenic protein expression, inactivates hormone-sensitive lipase (Hsl), and suppresses lipolysis. Conversely, mice that lack the neuronal insulin receptor exhibit unrestrained lipolysis and decreased de novo lipogenesis in WAT. Thus, brain and, in particular, hypothalamic insulin action play a pivotal role in WAT functionality.
Collapse
Affiliation(s)
- Thomas Scherer
- Department of Medicine, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1055, New York, NY 10029-6574, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|