1
|
Feng X, Ye Z, Xie K, Zhu S, Wu X, Sun Z, Feng X, Mo Y, Liang J, Shu G, Wang S, Zhu C, Jiang Q, Wang L. Effects of heat stress on the feeding preference of yellow-feathered broilers and its possible mechanism. J Therm Biol 2024; 124:103959. [PMID: 39180919 DOI: 10.1016/j.jtherbio.2024.103959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024]
Abstract
Heat stress is the most critical factor affecting animal feeding in summer. This experiment was conducted to investigate the effects of heat stress on the feeding preference of yellow-feathered broilers and its possible mechanism. As a result, the preference of yellow-feathered broilers for Tenebrio molitor was significantly decreased, and the fear response and serum corticosterone of broilers were significantly increased when the ambient temperatures are 35 °C (P < 0.05). In the central nervous system, consistent with the change in feeding preference, decreased dopamine in the nucleus accumbens (NAc) and increased mRNA levels of MAO-B in the ventral tegmental area (VTA) and NAc were found in yellow-feathered broilers (P < 0.05). In addition, we found significantly increased mRNA levels of corticotropin-releasing hormone receptor 1, corticotropin-releasing hormone receptor 2 and glucocorticoid receptor in the VTA and NAc of female broilers (P < 0.05). However, no similar change was found in male broilers. On the other hand, the serum levels of insulin and glucagon-like peptide-1 were increased only in male broilers (P < 0.05). Accordingly, the mRNA levels of insulin receptor and glucagon-like peptide-1 receptor in the VTA and the phosphorylation of mTOR and PI3K were increased only in male broilers (P < 0.05). In summary, the preference of yellow-feathered broilers for Tenebrio molitor feed decreased under heat stress conditions, and hedonic feeding behavior was significantly inhibited. However, the mechanism by which heat stress affects hedonic feeding behavior may contain gender differences. The insulin signaling pathway may participate in the regulation of heat stress on the male broiler reward system, while stress hormone-related receptors in the midbrain may play an important role in the effect of heat stress on the reward system of female broilers.
Collapse
Affiliation(s)
- Xiajie Feng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Ziyuan Ye
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Kailai Xie
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Shuqing Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Xin Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Zhonghua Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Xiaohua Feng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Yingfen Mo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Jingwen Liang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Gang Shu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Songbo Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Canjun Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Lina Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, 510642, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| |
Collapse
|
2
|
Antasouras G, Dakanalis A, Chrysafi M, Papadopoulou SK, Trifonidi I, Spanoudaki M, Alexatou O, Pritsa A, Louka A, Giaginis C. Could Insulin Be a Better Regulator of Appetite/Satiety Balance and Body Weight Maintenance in Response to Glucose Exposure Compared to Sucrose Substitutes? Unraveling Current Knowledge and Searching for More Appropriate Choices. Med Sci (Basel) 2024; 12:29. [PMID: 38921683 PMCID: PMC11205552 DOI: 10.3390/medsci12020029] [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: 04/13/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Insulin exerts a crucial impact on glucose control, cellular growing, function, and metabolism. It is partially modulated by nutrients, especially as a response to the intake of foods, including carbohydrates. Moreover, insulin can exert an anorexigenic effect when inserted into the hypothalamus of the brain, in which a complex network of an appetite/hunger control system occurs. The current literature review aims at thoroughly summarizing and scrutinizing whether insulin release in response to glucose exposure may be a better choice to control body weight gain and related diseases compared to the use of sucrose substitutes (SSs) in combination with a long-term, well-balanced diet. METHODS This is a comprehensive literature review, which was performed through searching in-depth for the most accurate scientific databases and applying effective and relevant keywords. RESULTS The insulin action can be inserted into the hypothalamic orexigenic/anorexigenic complex system, activating several anorexigenic peptides, increasing the hedonic aspect of food intake, and effectively controlling the human body weight. In contrast, SSs appear not to affect the orexigenic/anorexigenic complex system, resulting in more cases of uncontrolled body weight maintenance while also increasing the risk of developing related diseases. CONCLUSIONS Most evidence, mainly derived from in vitro and in vivo animal studies, has reinforced the insulin anorexigenic action in the hypothalamus of the brain. Simultaneously, most available clinical studies showed that SSs during a well-balanced diet either maintain or even increase body weight, which may indirectly be ascribed to the fact that they cannot cover the hedonic aspect of food intake. However, there is a strong demand for long-term longitudinal surveys to effectively specify the impact of SSs on human metabolic health.
Collapse
Affiliation(s)
- Georgios Antasouras
- Department of Food Science and Nutrition, School of Environment, University of Aegean, 81400 Lemnos, Greece; (G.A.); (M.C.); (O.A.); (A.L.)
| | - Antonios Dakanalis
- Department of Mental Health, Fondazione IRCCS San Gerardo dei Tintori, Via G.B. Pergolesi 33, 20900 Monza, Italy;
- Department of Medicine and Surgery, University of Milan Bicocca, Via Cadore 38, 20900 Monza, Italy
| | - Maria Chrysafi
- Department of Food Science and Nutrition, School of Environment, University of Aegean, 81400 Lemnos, Greece; (G.A.); (M.C.); (O.A.); (A.L.)
| | - Sousana K. Papadopoulou
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece; (S.K.P.); (M.S.); (A.P.)
| | - Ioulia Trifonidi
- Department of Clinical Biochemistry, KAT General Hospital, 14561 Athens, Greece;
| | - Maria Spanoudaki
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece; (S.K.P.); (M.S.); (A.P.)
| | - Olga Alexatou
- Department of Food Science and Nutrition, School of Environment, University of Aegean, 81400 Lemnos, Greece; (G.A.); (M.C.); (O.A.); (A.L.)
| | - Agathi Pritsa
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece; (S.K.P.); (M.S.); (A.P.)
| | - Aikaterini Louka
- Department of Food Science and Nutrition, School of Environment, University of Aegean, 81400 Lemnos, Greece; (G.A.); (M.C.); (O.A.); (A.L.)
| | - Constantinos Giaginis
- Department of Food Science and Nutrition, School of Environment, University of Aegean, 81400 Lemnos, Greece; (G.A.); (M.C.); (O.A.); (A.L.)
| |
Collapse
|
3
|
Bian H, Shao X, Cai W, Fu H. Understanding the Reversible Binding of a Multichain Protein-Protein Complex through Free-Energy Calculations. J Phys Chem B 2024; 128:3598-3604. [PMID: 38574232 DOI: 10.1021/acs.jpcb.4c00519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
We demonstrate that the binding affinity of a multichain protein-protein complex, insulin dimer, can be accurately predicted using a streamlined route of standard binding free-energy calculations. We find that chains A and C, which do not interact directly during binding, stabilize the insulin monomer structures and reduce the binding affinity of the two monomers, therefore enabling their reversible association. Notably, we confirm that although classical methods can estimate the binding affinity of the insulin dimer, conventional molecular dynamics, enhanced sampling algorithms, and classical geometrical routes of binding free-energy calculations may not fully capture certain aspects of the role played by the noninteracting chains in the binding dynamics. Therefore, this study not only elucidates the role of noninteracting chains in the reversible binding of the insulin dimer but also offers a methodological guide for investigating the reversible binding of multichain protein-protein complexes utilizing streamlined free-energy calculations.
Collapse
Affiliation(s)
- Hengwei Bian
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| | - Xueguang Shao
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
- School of Materials Science and Engineering, Smart Sensing Interdisciplinary Science Center, Nankai University, Tianjin 300350, China
| | - Wensheng Cai
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
- School of Materials Science and Engineering, Smart Sensing Interdisciplinary Science Center, Nankai University, Tianjin 300350, China
| | - Haohao Fu
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
- School of Materials Science and Engineering, Smart Sensing Interdisciplinary Science Center, Nankai University, Tianjin 300350, China
| |
Collapse
|
4
|
Basatinya AM, Sajedianfard J, Nazifi S, Hosseinzadeh S. The analgesic effects of insulin and its disorders in streptozotocin-induced short-term diabetes. Physiol Rep 2024; 12:e16009. [PMID: 38639646 PMCID: PMC11027902 DOI: 10.14814/phy2.16009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/01/2024] [Accepted: 04/01/2024] [Indexed: 04/20/2024] Open
Abstract
Evidence suggests that insulin resistance plays an important role in developing diabetes complications. The association between insulin resistance and pain perception is less well understood. This study aimed to investigate the effects of peripheral insulin deficiency on pain pathways in the brain. Diabetes was induced in 60 male rats with streptozotocin (STZ). Insulin was injected into the left ventricle of the brain by intracerebroventricular (ICV) injection, then pain was induced by subcutaneous injection of 2.5% formalin. Samples were collected at 4 weeks after STZ injection. Dopamine (DA), serotonin, reactive oxygen species (ROS), and mitochondrial glutathione (mGSH) were measured by ELISA, and gene factors were assessed by RT-qPCR. In diabetic rats, the levels of DA, serotonin, and mGSH decreased in the nuclei of the thalamus, raphe magnus, and periaqueductal gray, and the levels of ROS increased. In addition, the levels of expression of the neuron-specific enolase and receptor for advanced glycation end genes increased, but the expression of glial fibrillary acidic protein expression was reduced. These results support the findings that insulin has an analgesic effect in non-diabetic rats, as demonstrated by the formalin test. ICV injection of insulin reduces pain sensation, but this was not observed in diabetic rats, which may be due to cell damage ameliorated by insulin.
Collapse
Affiliation(s)
| | - Javad Sajedianfard
- Department of Basic Sciences, School of Veterinary MedicineShiraz UniversityShirazIran
| | - Saeed Nazifi
- Department of Clinical Science, School of Veterinary MedicineShiraz UniversityShirazIran
| | - Saied Hosseinzadeh
- Department of Food Hygiene and Public Health, School of Veterinary MedicineShiraz UniversityShirazIran
| |
Collapse
|
5
|
Camlik G, Bilakaya B, Ozsoy Y, Degim IT. A new approach for the treatment of Alzheimer's disease: insulin-quantum dots. J Microencapsul 2024; 41:18-26. [PMID: 37966713 DOI: 10.1080/02652048.2023.2282968] [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: 06/12/2023] [Accepted: 11/09/2023] [Indexed: 11/16/2023]
Abstract
The potential use of insulin supplementation for Alzheimer's Disease (AD) was aimed to investigate and explore CQDs as an alternative delivery system. CQDs were produced by microwave and characterised. Insulin-loaded Ins-CQDs and in-situ Gel-Ins-CQDs were developed. The in vitro release kinetics, penetrations of insulin through excised sheep nasal mucosa were determined. Toxicity of CQDs were calculated on SH-SY5Y cells. The stability and usability of the prepared formulations were assessed. The insulin release from the solution was 70.75% after 3 hours, while it was 37.51% for in-situ Gel-Ins-CQDs. IC50 value was 52 µM. The mean particle diameters of Ins-CQDs and in-situ Gel-Ins-CQDs varied between 8.35 ± 0.19 to 8.75 ± 0.03 nm during a 6-month period. Zeta potentials ranged from -31.51 ± 1.39 to -24.43 ± 0.26 mV, and PDI values were between 9.8 ± 0.01 to 5.3 ± 3.2%(SD, n = 3) for Ins-CQDs and in-situ Gel-Ins-CQDs, respectively.Our results show that Gel-Ins-CQDs represented a controlled release over time and can be used for AD through the nasal route.
Collapse
Affiliation(s)
- Gamze Camlik
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Biruni University, Istanbul, Türkiye
| | - Besa Bilakaya
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Biruni University, Istanbul, Türkiye
| | - Yildiz Ozsoy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul University, Istanbul, Türkiye
| | - Ismail Tuncer Degim
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Biruni University, Istanbul, Türkiye
| |
Collapse
|
6
|
Carr ST, Saito ER, Walton CM, Saito JY, Hanegan CM, Warren CE, Trumbull AM, Bikman BT. Ceramides Mediate Insulin-Induced Impairments in Cerebral Mitochondrial Bioenergetics in ApoE4 Mice. Int J Mol Sci 2023; 24:16635. [PMID: 38068958 PMCID: PMC10706658 DOI: 10.3390/ijms242316635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/13/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of neurodegenerative disease worldwide. A large body of work implicates insulin resistance in the development and progression of AD. Moreover, impairment in mitochondrial function, a common symptom of insulin resistance, now represents a fundamental aspect of AD pathobiology. Ceramides are a class of bioactive sphingolipids that have been hypothesized to drive insulin resistance. Here, we describe preliminary work that tests the hypothesis that hyperinsulinemia pathologically alters cerebral mitochondrial function in AD mice via accrual of the ceramides. Homozygous male and female ApoE4 mice, an oft-used model of AD research, were given chronic injections of PBS (control), insulin, myriocin (an inhibitor of ceramide biosynthesis), or insulin and myriocin over four weeks. Cerebral ceramide content was assessed using liquid chromatography-mass spectrometry. Mitochondrial oxygen consumption rates were measured with high-resolution respirometry, and H2O2 emissions were quantified via biochemical assays on brain tissue from the cerebral cortex. Significant increases in brain ceramides and impairments in brain oxygen consumption were observed in the insulin-treated group. These hyperinsulinemia-induced impairments in mitochondrial function were reversed with the administration of myriocin. Altogether, these data demonstrate a causative role for insulin in promoting brain ceramide accrual and subsequent mitochondrial impairments that may be involved in AD expression and progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Benjamin T. Bikman
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA; (S.T.C.); (E.R.S.); (C.M.W.); (A.M.T.)
| |
Collapse
|
7
|
Al-Kuraishy HM, Al-Gareeb AI, Alsayegh AA, Hakami ZH, Khamjan NA, Saad HM, Batiha GES, De Waard M. A Potential Link Between Visceral Obesity and Risk of Alzheimer's Disease. Neurochem Res 2023; 48:745-766. [PMID: 36409447 DOI: 10.1007/s11064-022-03817-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/05/2022] [Accepted: 11/08/2022] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia characterized by the deposition of amyloid beta (Aβ) plaques and tau-neurofibrillary tangles in the brain. Visceral obesity (VO) is usually associated with low-grade inflammation due to higher expression of pro-inflammatory cytokines by adipose tissue. The objective of the present review was to evaluate the potential link between VO and the development of AD. Tissue hypoxia in obesity promotes tissue injury, production of adipocytokines, and release of pro-inflammatory cytokines leading to an oxidative-inflammatory loop with induction of insulin resistance. Importantly, brain insulin signaling is involved in the pathogenesis of AD and lower cognitive function. Obesity and enlargement of visceral adipose tissue are associated with the deposition of Aβ. All of this is consonant with VO increasing the risk of AD through the dysregulation of adipocytokines which affect the development of AD. The activated nuclear factor kappa B (NF-κB) pathway in VO might be a potential link in the development of AD. Likewise, the higher concentration of advanced glycation end-products in VO could be implicated in the pathogenesis of AD. Taken together, different inflammatory signaling pathways are activated in VO that all have a negative impact on the cognitive function and progression of AD except hypoxia-inducible factor 1 which has beneficial and neuroprotective effects in mitigating the progression of AD. In addition, VO-mediated hypoadiponectinemia and leptin resistance may promote the progression of Aβ formation and tau phosphorylation with the development of AD. In conclusion, VO-induced AD is mainly mediated through the induction of oxidative stress, inflammatory changes, leptin resistance, and hypoadiponectinemia that collectively trigger Aβ formation and neuroinflammation. Thus, early recognition of VO by visceral adiposity index with appropriate management could be a preventive measure against the development of AD in patients with VO.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Abdulrahman A Alsayegh
- Clinical Nutrition Department, Applied Medical Sciences College, Jazan University, Jazan, 82817, Saudi Arabia
| | - Zaki H Hakami
- Medical Laboratory Technology Department Applied Medical Sciences College, Jazan University, Jazan, 82817, Saudi Arabia
| | - Nizar A Khamjan
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Egypt.
| | - Michel De Waard
- Smartox Biotechnology, 6 rue des Platanes, 38120, Saint-Egrève, France.,L'institut du thorax, INSERM, CNRS, UNIV NANTES, 44007, Nantes, France.,LabEx «Ion Channels, Science & Therapeutics», Université de Nice Sophia-Antipolis, 06560, Valbonne, France
| |
Collapse
|
8
|
Shpakov AO, Zorina II, Derkach KV. Hot Spots for the Use of Intranasal Insulin: Cerebral Ischemia, Brain Injury, Diabetes Mellitus, Endocrine Disorders and Postoperative Delirium. Int J Mol Sci 2023; 24:3278. [PMID: 36834685 PMCID: PMC9962062 DOI: 10.3390/ijms24043278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
A decrease in the activity of the insulin signaling system of the brain, due to both central insulin resistance and insulin deficiency, leads to neurodegeneration and impaired regulation of appetite, metabolism, endocrine functions. This is due to the neuroprotective properties of brain insulin and its leading role in maintaining glucose homeostasis in the brain, as well as in the regulation of the brain signaling network responsible for the functioning of the nervous, endocrine, and other systems. One of the approaches to restore the activity of the insulin system of the brain is the use of intranasally administered insulin (INI). Currently, INI is being considered as a promising drug to treat Alzheimer's disease and mild cognitive impairment. The clinical application of INI is being developed for the treatment of other neurodegenerative diseases and improve cognitive abilities in stress, overwork, and depression. At the same time, much attention has recently been paid to the prospects of using INI for the treatment of cerebral ischemia, traumatic brain injuries, and postoperative delirium (after anesthesia), as well as diabetes mellitus and its complications, including dysfunctions in the gonadal and thyroid axes. This review is devoted to the prospects and current trends in the use of INI for the treatment of these diseases, which, although differing in etiology and pathogenesis, are characterized by impaired insulin signaling in the brain.
Collapse
Affiliation(s)
- Alexander O. Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| | | | | |
Collapse
|
9
|
Contreras CM, Gutiérrez-García AG. Insulin and fluoxetine produce opposite actions on lateral septal nucleus-infralimbic region connection responsivity. Behav Brain Res 2023; 437:114146. [PMID: 36202146 DOI: 10.1016/j.bbr.2022.114146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 09/28/2022] [Accepted: 10/02/2022] [Indexed: 11/07/2022]
Abstract
Some diabetes patients develop depression, the main treatment for which is antidepressants. Pharmacological interactions between insulin and antidepressants (e.g., fluoxetine) are controversial in the literature. Some authors reported hypoglycemic actions of fluoxetine, whereas others reported antidepressant-like actions. In healthy rats, insulin produces an antidespair-like action in rats through an increase in locomotor and exploratory activity, but differences in actions of insulin and fluoxetine on neuronal activity are unknown. The present study evaluated Wistar healthy rats that were treated with saline, insulin, fluoxetine, or fluoxetine + insulin for 3 days (short-term) or 21 days (long-term). The model consisted of electrical stimulation of the lateral septal nucleus (LSN) while we performed single-unit extracellular response recordings in the prelimbic cortex (PL) and infralimbic cortex (IL) subregions of the medial prefrontal cortex (mPFC). Stimulation of the LSN produced an initial brief excitatory paucisynaptic response and then a long-lasting inhibitory afterdischarge in the PL and IL. Treatment with saline and fluoxetine, but not insulin, minimally affected the paucisynaptic response. Differences were found in LSN-IL responsivity. The inhibitory afterdischarge was clearly enhanced in the long-term fluoxetine group but not by insulin alone or fluoxetine + insulin. These findings suggest that insulin produces some actions that are opposite to fluoxetine on LSN-mPFC connection responsivity, with no synergistic actions between the actions of insulin and fluoxetine.
Collapse
Affiliation(s)
- Carlos M Contreras
- Unidad Periférica del Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Xalapa, Veracruz 91190, Mexico.
| | - Ana G Gutiérrez-García
- Laboratorio de Neurofarmacología, Instituto de Neuroetología, Universidad Veracruzana, Xalapa, Veracruz 91190, Mexico
| |
Collapse
|
10
|
Davoudi P, Do DN, Colombo SM, Rathgeber B, Miar Y. Application of Genetic, Genomic and Biological Pathways in Improvement of Swine Feed Efficiency. Front Genet 2022; 13:903733. [PMID: 35754793 PMCID: PMC9220306 DOI: 10.3389/fgene.2022.903733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/20/2022] [Indexed: 12/24/2022] Open
Abstract
Despite the significant improvement of feed efficiency (FE) in pigs over the past decades, feed costs remain a major challenge for producers profitability. Improving FE is a top priority for the global swine industry. A deeper understanding of the biology underlying FE is crucial for making progress in genetic improvement of FE traits. This review comprehensively discusses the topics related to the FE in pigs including: measurements, genetics, genomics, biological pathways and the advanced technologies and methods involved in FE improvement. We first provide an update of heritability for different FE indicators and then characterize the correlations of FE traits with other economically important traits. Moreover, we present the quantitative trait loci (QTL) and possible candidate genes associated with FE in pigs and outline the most important biological pathways related to the FE traits in pigs. Finally, we present possible ways to improve FE in swine including the implementation of genomic selection, new technologies for measuring the FE traits, and the potential use of genome editing and omics technologies.
Collapse
Affiliation(s)
- Pourya Davoudi
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Duy Ngoc Do
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Stefanie M Colombo
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Bruce Rathgeber
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Younes Miar
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| |
Collapse
|
11
|
Takanche JS, Kim JE, Jang S, Yi HK. Insulin growth factor binding protein-3 enhances dental implant osseointegration against methylglyoxal-induced bone deterioration in a rat model. J Periodontal Implant Sci 2022; 52:155-169. [PMID: 35505576 PMCID: PMC9064780 DOI: 10.5051/jpis.2101200060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 11/08/2022] Open
Abstract
PURPOSE The aim of this study was to determine the effect of insulin growth factor binding protein-3 (IGFBP-3) on the inhibition of glucose oxidative stress and promotion of bone formation near the implant site in a rat model of methylglyoxal (MGO)-induced bone loss. METHODS An in vitro study was performed in MC3T3 E1 cells treated with chitosan gold nanoparticles (Ch-GNPs) conjugated with IGFBP-3 cDNA followed by MGO. An in vivo study was conducted in a rat model induced by MGO administration after the insertion of a dental implant coated with IGFBP-3. RESULTS MGO treatment downregulated molecules involved in osteogenic differentiation and bone formation in MC3T3 E1 cells and influenced the bone mineral density and bone volume of the femur and alveolar bone. In contrast, IGFBP-3 inhibited oxidative stress and inflammation and enhanced osteogenesis in MGO-treated MC3T3 E1 cells. In addition, IGFBP-3 promoted bone formation by reducing inflammatory proteins in MGO-administered rats. The application of Ch-GNPs conjugated with IGFBP-3 as a coating of titanium implants enhanced osteogenesis and the osseointegration of dental implants. CONCLUSIONS This study demonstrated that IGFBP-3 could be applied as a therapeutic component in dental implants to promote the osseointegration of dental implants in patients with diabetes, which affects MGO levels.
Collapse
Affiliation(s)
- Jyoti Shrestha Takanche
- Department of Oral Biochemistry, Institute of Oral Bioscience, School of Dentistry, Jeonbuk National University, Jeonju, Korea.,Department of Biochemistry, School of Medical Sciences, Kathmandu University, Nepal
| | - Ji-Eun Kim
- Department of Oral Biochemistry, Institute of Oral Bioscience, School of Dentistry, Jeonbuk National University, Jeonju, Korea
| | - Sungil Jang
- Department of Oral Biochemistry, Institute of Oral Bioscience, School of Dentistry, Jeonbuk National University, Jeonju, Korea
| | - Ho-Keun Yi
- Department of Oral Biochemistry, Institute of Oral Bioscience, School of Dentistry, Jeonbuk National University, Jeonju, Korea.
| |
Collapse
|
12
|
Gutiérrez-García AG, Contreras CM. Putative Anti-Immobility Action of Acute Insulin Is Attributable to an Increase in Locomotor Activity in Healthy Wistar Rats. Neuropsychobiology 2022; 80:483-492. [PMID: 33827082 DOI: 10.1159/000515141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/09/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Anti-immobility actions of insulin in diabetic rats that are subjected to the forced swim test (FST) have been reported. In this test, low doses of antidepressants exert actions after long-term treatment, without affecting locomotor activity in healthy rats. Few studies have compared acute and chronic actions of insulin with antidepressants in healthy rats. METHODS We hypothesized that if insulin exerts a true anti-immobility action, then its effects must be comparable to fluoxetine in both a 1-day treatment regimen and a 21-day treatment regimen in healthy, gonadally intact female Wistar rats. RESULTS The results showed that low levels of glycemia were produced by all treatments, including fluoxetine, and glycemia was lower in proestrus-estrus than in diestrus-metestrus. None of the treatments or regimens produced actions on indicators of anxiety in the elevated plus maze. Insulin in the 1-day regimen increased the number of crossings and rearings in the open field test and caused a low cumulative immobility time in the FST. These actions disappeared in the 21-day regimen. Compared with the other treatments, fluoxetine treatment alone or combined with insulin produced a longer latency to the first period of immobility and a shorter immobility time in the chronic regimen in the FST, without affecting locomotor activity, and more pronounced actions were observed in proestrus-estrus (i.e., a true anti-immobility effect). CONCLUSION These results indicate that insulin does not produce a true antidepressant action in healthy rats. The purported antidepressant effects that were observed were instead attributable to an increase in locomotor activity only in the 1-day regimen.
Collapse
Affiliation(s)
- Ana G Gutiérrez-García
- Instituto de Neuroetología, Laboratorio de Neurofarmacología, Universidad Veracruzana, Xalapa, Mexico
| | - Carlos M Contreras
- Instituto de Neuroetología, Laboratorio de Neurofarmacología, Universidad Veracruzana, Xalapa, Mexico.,Instituto de Investigaciones Biomédicas, Unidad Periférica Xalapa, Universidad Nacional Autónoma de México, Xalapa, Mexico
| |
Collapse
|
13
|
Internal state effects on behavioral shifts in freely behaving praying mantises (Tenodera sinensis). PLoS Comput Biol 2021; 17:e1009618. [PMID: 34928939 PMCID: PMC8751982 DOI: 10.1371/journal.pcbi.1009618] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 01/11/2022] [Accepted: 11/05/2021] [Indexed: 11/19/2022] Open
Abstract
How we interact with our environment largely depends on both the external cues presented by our surroundings and the internal state from within. Internal states are the ever-changing physiological conditions that communicate the immediate survival needs and motivate the animal to behaviorally fulfill them. Satiety level constitutes such a state, and therefore has a dynamic influence on the output behaviors of an animal. In predatory insects like the praying mantis, hunting tactics, grooming, and mating have been shown to change hierarchical organization of behaviors depending on satiety. Here, we analyze behavior sequences of freely hunting praying mantises (Tenodera sinensis) to explore potential differences in sequential patterning of behavior as a correlate of satiety. First, our data supports previous work that showed starved praying mantises were not just more often attentive to prey, but also more often attentive to further prey. This was indicated by the increased time fraction spent in attentive bouts such as prey monitoring, head turns (to track prey), translations (closing the distance to the prey), and more strike attempts. With increasing satiety, praying mantises showed reduced time in these behaviors and exhibited them primarily towards close-proximity prey. Furthermore, our data demonstrates that during states of starvation, the praying mantis exhibits a stereotyped pattern of behavior that is highly motivated by prey capture. As satiety increased, the sequenced behaviors became more variable, indicating a shift away from the necessity of prey capture to more fluid presentations of behavior assembly.
Collapse
|
14
|
Shinjyo N, Kita K. Infection and Immunometabolism in the Central Nervous System: A Possible Mechanistic Link Between Metabolic Imbalance and Dementia. Front Cell Neurosci 2021; 15:765217. [PMID: 34795562 PMCID: PMC8592913 DOI: 10.3389/fncel.2021.765217] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Metabolic syndromes are frequently associated with dementia, suggesting that the dysregulation of energy metabolism can increase the risk of neurodegeneration and cognitive impairment. In addition, growing evidence suggests the link between infections and brain disorders, including Alzheimer's disease. The immune system and energy metabolism are in an intricate relationship. Infection triggers immune responses, which are accompanied by imbalance in cellular and organismal energy metabolism, while metabolic disorders can lead to immune dysregulation and higher infection susceptibility. In the brain, the activities of brain-resident immune cells, including microglia, are associated with their metabolic signatures, which may be affected by central nervous system (CNS) infection. Conversely, metabolic dysregulation can compromise innate immunity in the brain, leading to enhanced CNS infection susceptibility. Thus, infection and metabolic imbalance can be intertwined to each other in the etiology of brain disorders, including dementia. Insulin and leptin play pivotal roles in the regulation of immunometabolism in the CNS and periphery, and dysfunction of these signaling pathways are associated with cognitive impairment. Meanwhile, infectious complications are often comorbid with diabetes and obesity, which are characterized by insulin resistance and leptin signaling deficiency. Examples include human immunodeficiency virus (HIV) infection and periodontal disease caused by an oral pathogen Porphyromonas gingivalis. This review explores potential interactions between infectious agents and insulin and leptin signaling pathways, and discuss possible mechanisms underlying the relationship between infection, metabolic dysregulation, and brain disorders, particularly focusing on the roles of insulin and leptin.
Collapse
Affiliation(s)
- Noriko Shinjyo
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan.,Laboratory of Immune Homeostasis, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Kiyoshi Kita
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan.,Department of Host-Defense Biochemistry, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
15
|
Gley K, Hadlich F, Trakooljul N, Haack F, Murani E, Gimsa U, Wimmers K, Ponsuksili S. Multi-Transcript Level Profiling Revealed Distinct mRNA, miRNA, and tRNA-Derived Fragment Bio-Signatures for Coping Behavior Linked Haplotypes in HPA Axis and Limbic System. Front Genet 2021; 12:635794. [PMID: 34490028 PMCID: PMC8417057 DOI: 10.3389/fgene.2021.635794] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 08/03/2021] [Indexed: 01/10/2023] Open
Abstract
The molecular basis of porcine coping behavior (CB) relies on a sophisticated interplay of genetic and epigenetic features. Deep sequencing technologies allowed the identification of a plethora of new regulatory small non-coding RNA (sncRNA). We characterized mRNA and sncRNA profiles of central parts of the physiological stress response system including amygdala, hippocampus, hypothalamus and adrenal gland using systems biology for integration. Therefore, ten each of high- (HR) and low- (LR) reactive pigs (n = 20) carrying a CB associated haplotype in a prominent QTL-region on SSC12 were selected for mRNA and sncRNA expression profiling. The molecular markers related to the LR group included ATP1B2, MPDU1, miR-19b-5p, let-7g-5p, and 5′-tiRNALeu in the adrenal gland, miR-194a-5p, miR-125a-5p, miR-7-1-5p, and miR-107-5p in the hippocampus and CBL and PVRL1 in the hypothalamus. Interestingly, amygdalae of the LR group showed 5′-tiRNA and 5′-tRF (5′-tRFLys, 5′-tiRNALys, 5′-tiRNACys, and 5′-tiRNAGln) enrichment. Contrarily, molecular markers associated with the HR group encompassed miR-26b-5p, tRNAArg, tRNAGlyiF in the adrenal gland, IGF1 and APOD in the amygdala and PBX1, TOB1, and C18orf1 in the hippocampus and miR-24 in the hypothalamus. In addition, hypothalami of the HR group were characterized by 3′-tiRNA enrichment (3′-tiRNAGln, 3′-tiRNAAsn, 3′-tiRNAVal, 3′-tRFPro, 3′-tiRNACys, and 3′-tiRNAAla) and 3′-tRFs enrichment (3′-tRFAsn, 3′-tRFGlu, and 3′-tRFVal). These evidence suggest that tRNA-derived fragments and their cleavage activity are a specific marker for coping behavior. Data integration revealed new bio-signatures of important molecular interactions on a multi-transcript level in HPA axis and limbic system of pigs carrying a CB-associated haplotype.
Collapse
Affiliation(s)
- Kevin Gley
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Dummerstorf, Germany
| | - Frieder Hadlich
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Dummerstorf, Germany
| | - Nares Trakooljul
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Dummerstorf, Germany
| | - Fiete Haack
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Dummerstorf, Germany
| | - Eduard Murani
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Dummerstorf, Germany
| | - Ulrike Gimsa
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Behavioral Physiology, Dummerstorf, Germany
| | - Klaus Wimmers
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Dummerstorf, Germany
| | - Siriluck Ponsuksili
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Dummerstorf, Germany
| |
Collapse
|
16
|
Khatri DK, Kadbhane A, Patel M, Nene S, Atmakuri S, Srivastava S, Singh SB. Gauging the role and impact of drug interactions and repurposing in neurodegenerative disorders. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100022. [PMID: 34909657 PMCID: PMC8663985 DOI: 10.1016/j.crphar.2021.100022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/23/2021] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases (ND) are of vast origin which are characterized by gradual progressive loss of neurons in the brain region. ND can be classified according to the clinical symptoms present (e.g. Cognitive decline, hyperkinetic, and hypokinetic movements disorder) or by the pathological protein deposited (e.g., Amyloid, tau, Alpha-synuclein, TDP-43). Alzheimer's disease preceded by Parkinson's is the most prevalent form of ND world-wide. Multiple factors like aging, genetic mutations, environmental factors, gut microbiota, blood-brain barrier microvascular complication, etc. may increase the predisposition towards ND. Genetic mutation is a major contributor in increasing the susceptibility towards ND, the concept of one disease-one gene is obsolete and now multiple genes are considered to be involved in causing one particular disease. Also, the involvement of multiple pathological mechanisms like oxidative stress, neuroinflammation, mitochondrial dysfunction, etc. contributes to the complexity and makes them difficult to be treated by traditional mono-targeted ligands. In this aspect, the Poly-pharmacological drug approach which targets multiple pathological pathways at the same time provides the best way to treat such complex networked CNS diseases. In this review, we have provided an overview of ND and their pathological origin, along with a brief description of various genes associated with multiple diseases like Alzheimer's, Parkinson's, Multiple sclerosis (MS), Amyotrophic Lateral Sclerosis (ALS), Huntington's and a comprehensive detail about the Poly-pharmacology approach (MTDLs and Fixed-dose combinations) along with their merits over the traditional single-targeted drug is provided. This review also provides insights into current repurposing strategies along with its regulatory considerations.
Collapse
Affiliation(s)
- Dharmendra Kumar Khatri
- Corresponding authors. Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India.
| | | | | | | | | | | | - Shashi Bala Singh
- Corresponding authors. Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India.
| |
Collapse
|
17
|
Yu J, Liao X, Zhong Y, Wu Y, Lai X, Jiao H, Yan M, Zhang Y, Ma C, Wang S. The Candidate Schizophrenia Risk Gene Tmem108 Regulates Glucose Metabolism Homeostasis. Front Endocrinol (Lausanne) 2021; 12:770145. [PMID: 34690937 PMCID: PMC8531597 DOI: 10.3389/fendo.2021.770145] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 09/23/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Schizophrenia (SCZ) is a severe psychiatric disease affected by genetic factors and environmental contributors, and premorbid abnormality of glucose metabolism is one of the SCZ characteristics supposed to contribute to the disease's pathological process. Transmembrane protein 108 (Tmem108) is a susceptible gene associated with multiple psychiatric diseases, including SCZ. Moreover, Tmem108 mutant mice exhibit SCZ-like behaviors in the measurement of sensorimotor gating. However, it is unknown whether Tmem108 regulates glucose metabolism homeostasis while it involves SCZ pathophysiological process. RESULTS In this research, we found that Tmem108 mutant mice exhibited glucose intolerance, insulin resistance, and disturbed metabolic homeostasis. Food and oxygen consumption decreased, and urine production increased, accompanied by weak fatigue resistance in the mutant mice. Simultaneously, the glucose metabolic pathway was enhanced, and lipid metabolism decreased in the mutant mice, consistent with the elevated respiratory exchange ratio (RER). Furthermore, metformin attenuated plasma glucose levels and improved sensorimotor gating in Tmem108 mutant mice. CONCLUSIONS Hyperglycemia occurs more often in SCZ patients than in control, implying that these two diseases share common biological mechanisms, here we demonstrate that the Tmem108 mutant may represent such a comorbid mechanism.
Collapse
Affiliation(s)
- Jianbo Yu
- Laboratory of Synaptic Development and Plasticity, Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang, China
| | - Xufeng Liao
- Laboratory of Synaptic Development and Plasticity, Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang, China
| | - Yanzi Zhong
- Laboratory of Synaptic Development and Plasticity, Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang, China
- Department of Biology, Senior Middle School of Yongfeng, Ji’an, China
| | - Yongqiang Wu
- Laboratory of Synaptic Development and Plasticity, Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang, China
| | - Xinsheng Lai
- Laboratory of Synaptic Development and Plasticity, Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang, China
- School of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Huifeng Jiao
- School of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Min Yan
- School of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Yu Zhang
- Laboratory of Synaptic Development and Plasticity, Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang, China
| | - Chaolin Ma
- Laboratory of Synaptic Development and Plasticity, Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang, China
- School of Basic Medical Sciences, Nanchang University, Nanchang, China
- *Correspondence: Chaolin Ma, ; Shunqi Wang,
| | - Shunqi Wang
- Laboratory of Synaptic Development and Plasticity, Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang, China
- School of Basic Medical Sciences, Nanchang University, Nanchang, China
- *Correspondence: Chaolin Ma, ; Shunqi Wang,
| |
Collapse
|
18
|
Xu C, Wang X, Zhou S, Wu J, Geng Q, Ruan D, Qiu Y, Quan J, Ding R, Cai G, Wu Z, Zheng E, Yang J. Brain Transcriptome Analysis Reveals Potential Transcription Factors and Biological Pathways Associated with Feed Efficiency in Commercial DLY Pigs. DNA Cell Biol 2020; 40:272-282. [PMID: 33297854 DOI: 10.1089/dna.2020.6071] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Feed efficiency (FE) is one of the most important economic traits in the porcine industry. In this study, high-throughput RNA sequencing (RNA-seq) was first utilized for brain tissue transcriptome analysis in pigs to indicate the potential genes and biological pathways related to FE in pigs. A total of 8 pigs with either extremely high-FE group (HE-group) or low-FE group (LE-group) were selected from 225 Duroc × (Landrace × Yorkshire) (DLY) pigs for transcriptomic analysis. RNA-seq analysis was performed to determine differentially expressed genes (DEGs) between the HE- and LE-group, and 430 DEGs were identified in brain tissues of pigs (|log2(FoldChange)| > 1; adjusted p-values <0.05). Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed that the DEGs were mainly enriched in synaptic signaling or transmission, and hormone secretion pathways, in which insulin secretion, and oxytocin signaling pathways were closely associated with FE by regulating feeding behavior and energy metabolism (adjusted p-values <0.05). Further, the transcription factors (TFs) analysis and gene co-expression network analysis indicated three hub differentially expressed TFs (NR2F2, TFAP2D, and HNF1B) that affected FE by mainly regulating feeding behavior, insulin sensitivity, or energy metabolism. Our findings suggest several potential TFs and biological pathways for further investigations of FE in pigs.
Collapse
Affiliation(s)
- Cineng Xu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Xingwang Wang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Shenping Zhou
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Jie Wu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Qian Geng
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Donglin Ruan
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Yibin Qiu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Jianping Quan
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Rongrong Ding
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Gengyuan Cai
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Zhenfang Wu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Enqin Zheng
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Jie Yang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| |
Collapse
|
19
|
Insulin Resistance in Osteoarthritis: Similar Mechanisms to Type 2 Diabetes Mellitus. J Nutr Metab 2020; 2020:4143802. [PMID: 32566279 PMCID: PMC7261331 DOI: 10.1155/2020/4143802] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/01/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) and type 2 diabetes mellitus (T2D) are two of the most widespread chronic diseases. OA and T2D have common epidemiologic traits, are considered heterogenic multifactorial pathologies that develop through the interaction of genetic and environmental factors, and have common risk factors. In addition, both of these diseases often manifest in a single patient. Despite differences in clinical manifestations, both diseases are characterized by disturbances in cellular metabolism and by an insulin-resistant state primarily associated with the production and utilization of energy. However, currently, the primary cause of OA development and progression is not clear. In addition, although OA is manifested as a joint disease, evidence has accumulated that it affects the whole body. As pathological insulin resistance is viewed as a driving force of T2D development, now, we present evidence that the molecular and cellular metabolic disturbances associated with OA are linked to an insulin-resistant state similar to T2D. Moreover, the alterations in cellular energy requirements associated with insulin resistance could affect many metabolic changes in the body that eventually result in pathology and could serve as a unified mechanism that also functions in many metabolic diseases. However, these issues have not been comprehensively described. Therefore, here, we discuss the basic molecular mechanisms underlying the pathological processes associated with the development of insulin resistance; the major inducers, regulators, and metabolic consequences of insulin resistance; and instruments for controlling insulin resistance as a new approach to therapy.
Collapse
|
20
|
Lu A, Watkins M, Li Q, Robinson SD, Concepcion GP, Yandell M, Weng Z, Olivera BM, Safavi-Hemami H, Fedosov AE. Transcriptomic Profiling Reveals Extraordinary Diversity of Venom Peptides in Unexplored Predatory Gastropods of the Genus Clavus. Genome Biol Evol 2020; 12:684-700. [PMID: 32333764 PMCID: PMC7259678 DOI: 10.1093/gbe/evaa083] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2020] [Indexed: 12/21/2022] Open
Abstract
Predatory gastropods of the superfamily Conoidea number over 12,000 living species. The evolutionary success of this lineage can be explained by the ability of conoideans to produce complex venoms for hunting, defense, and competitive interactions. Whereas venoms of cone snails (family Conidae) have become increasingly well studied, the venoms of most other conoidean lineages remain largely uncharacterized. In the present study, we present the venom gland transcriptomes of two species of the genus Clavus that belong to the family Drilliidae. Venom gland transcriptomes of two specimens of Clavus canalicularis and two specimens of Clavus davidgilmouri were analyzed, leading to the identification of a total of 1,176 putative venom peptide toxins (drillipeptides). Based on the combined evidence of secretion signal sequence identity, entire precursor similarity search (BLAST), and the orthology inference, putative Clavus toxins were assigned to 158 different gene families. The majority of identified transcripts comprise signal, pro-, mature peptide, and post-regions, with a typically short (<50 amino acids) and cysteine-rich mature peptide region. Thus, drillipeptides are structurally similar to conotoxins. However, convincing homology with known groups of Conus toxins was only detected for very few toxin families. Among these are Clavus counterparts of Conus venom insulins (drillinsulins), porins (drilliporins), and highly diversified lectins (drillilectins). The short size of most drillipeptides and structural similarity to conotoxins were unexpected, given that most related conoidean gastropod families (Terebridae and Turridae) possess longer mature peptide regions. Our findings indicate that, similar to conotoxins, drillipeptides may represent a valuable resource for future pharmacological exploration.
Collapse
Affiliation(s)
- Aiping Lu
- Department of Central Laboratory, Shanghai Tenth People’s Hospital of Tongji University, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | | | - Qing Li
- Eccles Institute of Human Genetics, University of Utah
- High-Throughput Genomics and Bioinformatic Analysis Shared Resource, Huntsman Cancer Institute, University of Utah
| | | | - Gisela P Concepcion
- Marine Science Institute, University of the Philippines-Diliman, Quezon City, Philippines
| | - Mark Yandell
- Eccles Institute of Human Genetics, University of Utah
- Utah Center for Genetic Discovery, University of Utah
| | - Zhiping Weng
- Department of Central Laboratory, Shanghai Tenth People’s Hospital of Tongji University, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School
| | | | - Helena Safavi-Hemami
- Department of Biochemistry, University of Utah
- Department of Biology, University of Copenhagen, Denmark
| | - Alexander E Fedosov
- A.N. Severtsov Institute of Ecology and Evolution, Russian Academy of Science, Moscow, Russia
| |
Collapse
|
21
|
Type 3 Diabetes and Its Role Implications in Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21093165. [PMID: 32365816 PMCID: PMC7246646 DOI: 10.3390/ijms21093165] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 04/21/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022] Open
Abstract
The exact connection between Alzheimer’s disease (AD) and type 2 diabetes is still in debate. However, poorly controlled blood sugar may increase the risk of developing Alzheimer’s. This relationship is so strong that some have called Alzheimer’s “diabetes of the brain” or “type 3 diabetes (T3D)”. Given more recent studies continue to indicate evidence linking T3D with AD, this review aims to demonstrate the relationship between T3D and AD based on the fact that both the processing of amyloid-β (Aβ) precursor protein toxicity and the clearance of Aβ are attributed to impaired insulin signaling, and that insulin resistance mediates the dysregulation of bioenergetics and progress to AD. Furthermore, insulin-related therapeutic strategies are suggested to succeed in the development of therapies for AD by slowing down their progressive nature or even halting their future complications.
Collapse
|
22
|
Dubey SK, Lakshmi KK, Krishna KV, Agrawal M, Singhvi G, Saha RN, Saraf S, Saraf S, Shukla R, Alexander A. Insulin mediated novel therapies for the treatment of Alzheimer's disease. Life Sci 2020; 249:117540. [PMID: 32165212 DOI: 10.1016/j.lfs.2020.117540] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/03/2020] [Accepted: 03/07/2020] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease, a progressive neurodegenerative disorder, is one of the leading causes of death in the USA, along with cancer and cardiac disorders. AD is characterized by various neurological factors like amyloid plaques, tau hyperphosphorylation, mitochondrial dysfunction, acetylcholine deficiency, etc. Together, impaired insulin signaling in the brain is also observed as essential factor to be considered in AD pathophysiology. Hence, currently researchers focused on studying the effect of brain insulin metabolism and relation of diabetes with AD. Based on the investigations, AD is also considered as type 3 or brain diabetes. Besides the traditional view of correlating AD with aging, a better understanding of various pathological factors and effects of other physical ailments is necessary to develop a promising therapeutic approach. There is a vast scope of studying the relation of systemic insulin level, insulin signaling, its neuroprotective potency and effect of diabetes on AD progression. The present work describes worldwide status of AD and its relation with diabetes mellitus and insulin metabolism; pathophysiology of AD; different metabolic pathways associating insulin metabolism with AD; insulin receptor and signaling in the brain; glucose metabolism; insulin resistance; and various preclinical and clinical studies reported insulin-based therapies to treat AD via systemic route and through direct intranasal delivery to the brain.
Collapse
Affiliation(s)
- Sunil Kumar Dubey
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Rajasthan, India.
| | - K K Lakshmi
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Rajasthan, India
| | - Kowthavarapu Venkata Krishna
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Rajasthan, India
| | - Mukta Agrawal
- Rungta College of Pharmaceutical Sciences and Research, Kohka-Kurud Road, Bhilai, Chhattisgarh 490 024, India
| | - Gautam Singhvi
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Rajasthan, India
| | - Ranendra Narayana Saha
- Department of Biotechnology, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Dubai Campus, Dubai, United Arab Emirates
| | - Swarnlata Saraf
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chhattisgarh 492010, India
| | - Shailendra Saraf
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chhattisgarh 492010, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER-R), New Transit Campus, Bijnor Road, Sarojini Nagar, Lucknow 226002, India
| | - Amit Alexander
- National Institute of Pharmaceutical Education and Research (NIPER-Guwahati), Department of Pharmaceuticals, Ministry of Chemicals & Fertilizers, Govt. of India, NH 37, NITS Mirza, Kamrup-781125, Guwahati, Assam, India.
| |
Collapse
|
23
|
McNay EC, Pearson-Leary J. GluT4: A central player in hippocampal memory and brain insulin resistance. Exp Neurol 2020; 323:113076. [PMID: 31614121 PMCID: PMC6936336 DOI: 10.1016/j.expneurol.2019.113076] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/19/2019] [Accepted: 10/01/2019] [Indexed: 12/24/2022]
Abstract
Insulin is now well-established as playing multiple roles within the brain, and specifically as regulating hippocampal cognitive processes and metabolism. Impairments to insulin signaling, such as those seen in type 2 diabetes and Alzheimer's disease, are associated with brain hypometabolism and cognitive impairment, but the mechanisms of insulin's central effects are not determined. Several lines of research converge to suggest that the insulin-responsive glucose transporter GluT4 plays a central role in hippocampal memory processes, and that reduced activation of this transporter may underpin the cognitive impairments seen as a consequence of insulin resistance.
Collapse
Affiliation(s)
- Ewan C McNay
- Behavioral Neuroscience, University at Albany, Albany, NY, USA.
| | - Jiah Pearson-Leary
- Department of Anesthesiology, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
24
|
Fiory F, Perruolo G, Cimmino I, Cabaro S, Pignalosa FC, Miele C, Beguinot F, Formisano P, Oriente F. The Relevance of Insulin Action in the Dopaminergic System. Front Neurosci 2019; 13:868. [PMID: 31474827 PMCID: PMC6706784 DOI: 10.3389/fnins.2019.00868] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 08/02/2019] [Indexed: 12/13/2022] Open
Abstract
The advances in medicine, together with lifestyle modifications, led to a rising life expectancy. Unfortunately, however, aging is accompanied by an alarming boost of age-associated chronic pathologies, including neurodegenerative and metabolic diseases. Interestingly, a non-negligible interplay between alterations of glucose homeostasis and brain dysfunction has clearly emerged. In particular, epidemiological studies have pointed out a possible association between Type 2 Diabetes (T2D) and Parkinson’s Disease (PD). Insulin resistance, one of the major hallmark for etiology of T2D, has a detrimental influence on PD, negatively affecting PD phenotype, accelerating its progression and worsening cognitive impairment. This review aims to provide an exhaustive analysis of the most recent evidences supporting the key role of insulin resistance in PD pathogenesis. It will focus on the relevance of insulin in the brain, working as pro-survival neurotrophic factor and as a master regulator of neuronal mitochondrial function and oxidative stress. Insulin action as a modulator of dopamine signaling and of alpha-synuclein degradation will be described in details, too. The intriguing idea that shared deregulated pathogenic pathways represent a link between PD and insulin resistance has clinical and therapeutic implications. Thus, ongoing studies about the promising healing potential of common antidiabetic drugs such as metformin, exenatide, DPP IV inhibitors, thiazolidinediones and bromocriptine, will be summarized and the rationale for their use to decelerate neurodegeneration will be critically assessed.
Collapse
Affiliation(s)
- Francesca Fiory
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT "Genomic of Diabetes," Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Giuseppe Perruolo
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT "Genomic of Diabetes," Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Ilaria Cimmino
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT "Genomic of Diabetes," Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Serena Cabaro
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT "Genomic of Diabetes," Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Francesca Chiara Pignalosa
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT "Genomic of Diabetes," Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Claudia Miele
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT "Genomic of Diabetes," Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Francesco Beguinot
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT "Genomic of Diabetes," Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Pietro Formisano
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT "Genomic of Diabetes," Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Francesco Oriente
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT "Genomic of Diabetes," Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| |
Collapse
|
25
|
Jarrett BY, Vantman N, Mergler RJ, Brooks ED, Pierson RA, Chizen DR, Lujan ME. Dysglycemia, Not Altered Sex Steroid Hormones, Affects Cognitive Function in Polycystic Ovary Syndrome. J Endocr Soc 2019; 3:1858-1868. [PMID: 31583367 PMCID: PMC6767628 DOI: 10.1210/js.2019-00112] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 07/24/2019] [Indexed: 02/07/2023] Open
Abstract
Context Polycystic ovary syndrome (PCOS) is a complex endocrine condition characterized by multiple reproductive and metabolic abnormalities. Because individual reproductive and metabolic abnormalities modulate working memory in the general population, there is growing interest in whether cognitive function is dually and negatively affected in PCOS. Objective To examine the association of reproductive and metabolic features with cognitive function in women with and without PCOS. Design An observational, cross-sectional study was conducted at an academic clinical research center in North America between 2006 and 2009. Common tests of working memory (i.e., manual dexterity, perceptual speed, and visuospatial ability) were performed by women with PCOS (n = 40) and control subjects (n = 40). Markers of sex steroid hormones, ovulatory function, and cardiometabolic health were also assessed. Results Reduced visuospatial ability was observed in women with PCOS compared with control subjects (P < 0.01). Reduced visuospatial ability was linked to higher levels of hemoglobin A1c in the entire study cohort, independent of body mass index or PCOS status. No associations were observed between visuospatial ability and reproductive features, after controlling for confounding variables. Conclusion Our findings support a role for glycemic control, and not PCOS per se, in cognitive dysfunction in women of reproductive age. Additional studies are needed to understand the short- and long-term effects of dysglycemia on brain health in women with PCOS, given their increased propensity for metabolic comorbidities, compared with control subjects.
Collapse
Affiliation(s)
| | - Natalie Vantman
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Reid J Mergler
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Eric D Brooks
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Roger A Pierson
- Department of Obstetrics and Gynecology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Donna R Chizen
- Department of Obstetrics and Gynecology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Marla E Lujan
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| |
Collapse
|
26
|
Jojo GM, Kuppusamy G, Selvaraj K, Baruah UK. Prospective of managing impaired brain insulin signalling in late onset Alzheimers disease with excisting diabetic drugs. J Diabetes Metab Disord 2019; 18:229-242. [PMID: 31275894 DOI: 10.1007/s40200-019-00405-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 04/12/2019] [Indexed: 12/25/2022]
Abstract
Late onset Alzheimer's disease (AD) is the most common cause of dementia among elderly. The exact cause of the disease is until now unknown and there is no complete cure for the disease. Growing evidence suggest that AD is a metabolic disorder associated with impairment in brain insulin signalling. These findings enriched the scope for the repurposing of diabetic drugs in AD management. Even though many of these drugs are moving in a positive direction in the ongoing clinical studies, the extent of the success has seen to influence by several properties of these drugs since they were originally designed to manage the peripheral insulin resistance. In depth understandings of these properties is hence highly significant to optimise the use of diabetic drugs in the clinical management of AD; which is the primary aim of the present review article.
Collapse
Affiliation(s)
- Gifty M Jojo
- Department of Pharmaceutics, JSS College of pharmacy, Ootacamund, JSS Academy of Higher Education & Research, Mysore, India
| | - Gowthamarajan Kuppusamy
- Department of Pharmaceutics, JSS College of pharmacy, Ootacamund, JSS Academy of Higher Education & Research, Mysore, India
| | - Kousalya Selvaraj
- Department of Pharmaceutics, JSS College of pharmacy, Ootacamund, JSS Academy of Higher Education & Research, Mysore, India
| | - Uday Krishna Baruah
- Department of Pharmaceutics, JSS College of pharmacy, Ootacamund, JSS Academy of Higher Education & Research, Mysore, India
| |
Collapse
|
27
|
Al-Zubaidi A, Heldmann M, Mertins A, Brabant G, Nolde JM, Jauch-Chara K, Münte TF. Impact of Hunger, Satiety, and Oral Glucose on the Association Between Insulin and Resting-State Human Brain Activity. Front Hum Neurosci 2019; 13:162. [PMID: 31178708 PMCID: PMC6544009 DOI: 10.3389/fnhum.2019.00162] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 05/01/2019] [Indexed: 12/12/2022] Open
Abstract
To study the interplay of metabolic state (hungry vs. satiated) and glucose administration (including hormonal modulation) on brain function, resting-state functional magnetic resonance imaging (rs-fMRI) and blood samples were obtained in 24 healthy normal-weight men in a repeated measurement design. Participants were measured twice: once after a 36 h fast (except water) and once under satiation (three meals/day for 36 h). During each session, rs-fMRI and hormone concentrations were recorded before and after a 75 g oral dose of glucose. We calculated the amplitude map from blood-oxygen-level-dependent (BOLD) signals by using the fractional amplitude of low-frequency fluctuation (fALFF) approach for each volunteer per condition. Using multiple linear regression analysis (MLRA) the interdependence of brain activity, plasma insulin and blood glucose was investigated. We observed a modulatory impact of fasting state on intrinsic brain activity in the posterior cingulate cortex (PCC). Strikingly, differences in plasma insulin levels between hunger and satiety states after glucose administration at the time of the scan were negatively related to brain activity in the posterior insula and superior frontal gyrus (SFG), while plasma glucose levels were positively associated with activity changes in the fusiform gyrus. Furthermore, we could show that changes in plasma insulin enhanced the connectivity between the posterior insula and SFG. Our results indicate that hormonal signals like insulin alleviate an acute hemostatic energy deficit by modifying the homeostatic and frontal circuitry of the human brain.
Collapse
Affiliation(s)
| | - Marcus Heldmann
- Department of Neurology, University of Lübeck, Lübeck, Germany
- Institute of Psychology II, University of Lübeck, Lübeck, Germany
| | - Alfred Mertins
- Institute for Signal Processing, University of Lübeck, Lübeck, Germany
| | - Georg Brabant
- Department of Internal Medicine I, University of Lübeck, Lübeck, Germany
| | | | - Kamila Jauch-Chara
- Department of Psychiatry and Psychotherapy, Christian-Albrechts-University, Kiel, Germany
| | - Thomas F. Münte
- Department of Neurology, University of Lübeck, Lübeck, Germany
- Institute of Psychology II, University of Lübeck, Lübeck, Germany
| |
Collapse
|
28
|
Lyra E Silva NDM, Gonçalves RA, Boehnke SE, Forny-Germano L, Munoz DP, De Felice FG. Understanding the link between insulin resistance and Alzheimer's disease: Insights from animal models. Exp Neurol 2019; 316:1-11. [PMID: 30930096 DOI: 10.1016/j.expneurol.2019.03.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease affecting millions of people worldwide. AD is characterized by a profound impairment of higher cognitive functions and still lacks any effective disease-modifying treatment. Defective insulin signaling has been implicated in AD pathophysiology, but the mechanisms underlying this process are not fully understood. Here, we review the molecular mechanisms underlying defective brain insulin signaling in rodent models of AD, and in a non-human primate (NHP) model of the disease that recapitulates features observed in AD brains. We further highlight similarities between the NHP and human brains and discuss why NHP models of AD are important to understand disease mechanisms and to improve the translation of effective therapies to humans. We discuss how studies using different animal models have contributed to elucidate the link between insulin resistance and AD.
Collapse
Affiliation(s)
| | | | - Susan E Boehnke
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Leticia Forny-Germano
- Institute of Medical Biochemistry Leopoldo De Meis, Federal University of Rio de Janeiro, Brazil
| | - Douglas P Munoz
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada.
| | - Fernanda G De Felice
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada; Department of Psychiatry, Queen's University, Kingston, ON, Canada; Institute of Medical Biochemistry Leopoldo De Meis, Federal University of Rio de Janeiro, Brazil.
| |
Collapse
|
29
|
Ahorukomeye P, Disotuar MM, Gajewiak J, Karanth S, Watkins M, Robinson SD, Flórez Salcedo P, Smith NA, Smith BJ, Schlegel A, Forbes BE, Olivera B, Hung-Chieh Chou D, Safavi-Hemami H. Fish-hunting cone snail venoms are a rich source of minimized ligands of the vertebrate insulin receptor. eLife 2019; 8:41574. [PMID: 30747102 PMCID: PMC6372279 DOI: 10.7554/elife.41574] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/30/2018] [Indexed: 12/27/2022] Open
Abstract
The fish-hunting marine cone snail Conus geographus uses a specialized venom insulin to induce hypoglycemic shock in its prey. We recently showed that this venom insulin, Con-Ins G1, has unique characteristics relevant to the design of new insulin therapeutics. Here, we show that fish-hunting cone snails provide a rich source of minimized ligands of the vertebrate insulin receptor. Insulins from C. geographus, Conus tulipa and Conus kinoshitai exhibit diverse sequences, yet all bind to and activate the human insulin receptor. Molecular dynamics reveal unique modes of action that are distinct from any other insulins known in nature. When tested in zebrafish and mice, venom insulins significantly lower blood glucose in the streptozotocin-induced model of diabetes. Our findings suggest that cone snails have evolved diverse strategies to activate the vertebrate insulin receptor and provide unique insight into the design of novel drugs for the treatment of diabetes. Insulin is a hormone critical for maintaining healthy blood sugar levels in humans. When the insulin system becomes faulty, blood sugar levels become too high, which can lead to diabetes. At the moment, the only effective treatment for one of the major types of diabetes are daily insulin injections. However, designing fast-acting insulin drugs has remained a challenge. Insulin molecules form clusters (so-called hexamers) that first have to dissolve in the body to activate the insulin receptor, which plays a key role in regulating the blood sugar levels throughout the body. This can take time and can therefore delay the blood-sugar control. In 2015, researchers discovered that the fish-hunting cone snail Conus geographus uses a specific type of insulin to capture its prey – fish. The cone snail releases insulin into the surrounding water and then engulfs its victim with its mouth. This induces dangerously low blood sugar levels in the fish and so makes them an easy target. Unlike the human version, the snail insulin does not cluster, and despite structural differences, can bind to the human insulin receptor. Now, Ahorukomeye, Disotuar et al. – including some of the authors involved in the previous study – wanted to find out whether other fish-hunting cone snails also make insulins and if they differed from the one previously discovered in C. geographus. The insulin molecules were extracted and analyzed, and the results showed that the three cone snail species had different versions of insulin – but none of them formed clusters. Ahorukomeye, Disotuar et al. further revealed that the snail insulins could bind to the human insulin receptors and could also reverse high blood sugar levels in fish and mouse models of the disease. This research may help guide future studies looking into developing fast-acting insulin drugs for diabetic patients. A next step will be to fully understand how snail insulins can be active at the human receptor without forming clusters. Cone snails solved this problem millions of years ago and by understanding how they have done this, researchers are hoping to redesign current diabetic therapeutics. Since the snail insulins do not form clusters and should act faster than currently available insulin drugs, they may lead to better or new diabetes treatments.
Collapse
Affiliation(s)
- Peter Ahorukomeye
- Department of Biology, University of Utah School of Medicine, Salt Lake City, United States
| | - Maria M Disotuar
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, United States
| | - Joanna Gajewiak
- Department of Biology, University of Utah School of Medicine, Salt Lake City, United States
| | - Santhosh Karanth
- Molecular Medicine Program, University of Utah, Salt Lake City, United States.,Department of Internal Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Utah School of Medicine, Salt Lake City, United States.,Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, United States
| | - Maren Watkins
- Department of Biology, University of Utah School of Medicine, Salt Lake City, United States
| | - Samuel D Robinson
- Department of Biology, University of Utah School of Medicine, Salt Lake City, United States
| | - Paula Flórez Salcedo
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, United States
| | - Nicholas A Smith
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Brian J Smith
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Amnon Schlegel
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, United States.,Molecular Medicine Program, University of Utah, Salt Lake City, United States.,Department of Internal Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Utah School of Medicine, Salt Lake City, United States.,Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, United States
| | - Briony E Forbes
- Department of Medical Biochemistry, Flinders University, Bedford Park, Australia
| | - Baldomero Olivera
- Department of Biology, University of Utah School of Medicine, Salt Lake City, United States
| | - Danny Hung-Chieh Chou
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, United States
| | - Helena Safavi-Hemami
- Department of Biology, University of Utah School of Medicine, Salt Lake City, United States.,Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, United States
| |
Collapse
|
30
|
Maghami S, Zardooz H, Khodagholi F, Binayi F, Ranjbar Saber R, Hedayati M, Sahraei H, Ansari MA. Maternal separation blunted spatial memory formation independent of peripheral and hippocampal insulin content in young adult male rats. PLoS One 2018; 13:e0204731. [PMID: 30332425 PMCID: PMC6192583 DOI: 10.1371/journal.pone.0204731] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 09/13/2018] [Indexed: 01/22/2023] Open
Abstract
This study explores the effects of maternal separation as a chronic early life stress (ELS) on pancreatic islets insulin content and secretion, and their potential relationship with the hippocampus insulin content and spatial memory in young adulthood. Male rat offspring were divided into two groups: stress (STR) and non-stress (non-STR) groups. The animals of the STR group were separated from their mothers during postnatal days (PND) 1 to 21. During the weaning time, that is, PND-0 to PND-21, the body weight and length of the pups were measured. Blood samples were collected on PND-1, 21, 29 and 34 and during young adulthood (53±2 days) to determine plasma corticosterone and insulin levels. The young adult animals were also tested for spatial memory. One day after the memory test, the animals were decapitated and their pancreases were removed to measure the islets insulin content and secretion. Finally, the animals' hippocampi were isolated to determine their insulin content and insulin receptor protein amounts. During the period of weaning, the body weight and length of pups belonging to the STR group were significantly lower as compared to those in the non-STR group. Maternal separation did not change the plasma levels of insulin but increased plasma corticosterone levels from PND-21 to young adulthood and also reduced the islets insulin content but did not affect insulin secretion and the hippocampus insulin content and insulin receptor protein amount. Although, at the end of the memory tests, rats of the STR group reached the escape box at almost the same time and distance and with the same errors as rats of the non-STR group, the distance traveled to reach the escape box showed a steep reduction in the non-STR group as compared to the STR group after the first trial. Moreover, as compared to the STR group, the non-STR group showed an increasing trend for direct strategy to find the escape box. The islets insulin content and secretion, and the plasma insulin concentration were not significantly correlated with the hippocampus insulin content. From the results of the present study, it appears that the main behavioral effect of the maternal separation stress in the spatial memory task was to impair the strategy used by the animals to reach the escape box. This may indicate that maternal separation stress affects brain regions other than the hippocampus. Moreover, due to the reduction of the body weight and length of offspring belonging to the STR group, it should be further considered that both maternal separation and early life malnutrition are directly (and mechanistically) linked to cognitive alterations later in life in ways that are not dependent on peripheral and hippocampal insulin content.
Collapse
Affiliation(s)
- Soheila Maghami
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homeira Zardooz
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- * E-mail: ,
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Binayi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roya Ranjbar Saber
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hedayat Sahraei
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Ansari
- Laser and Plasma Research Institute, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
31
|
Lázár BA, Jancsó G, Pálvölgyi L, Dobos I, Nagy I, Sántha P. Insulin Confers Differing Effects on Neurite Outgrowth in Separate Populations of Cultured Dorsal Root Ganglion Neurons: The Role of the Insulin Receptor. Front Neurosci 2018; 12:732. [PMID: 30364236 PMCID: PMC6191510 DOI: 10.3389/fnins.2018.00732] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 09/24/2018] [Indexed: 01/04/2023] Open
Abstract
Apart from its pivotal role in the regulation of carbohydrate metabolism, insulin exerts important neurotrophic and neuromodulator effects on dorsal root ganglion (DRG) neurons. The neurite outgrowth-promoting effect is one of the salient features of insulin's action on cultured DRG neurons. Although it has been established that a significant population of DRG neurons express the insulin receptor (InsR), the significance of InsR expression and the chemical phenotype of DRG neurons in relation to the neurite outgrowth-promoting effect of insulin has not been studied. Therefore, in this study by using immunohistochemical and quantitative stereological methods we evaluated the effect of insulin on neurite outgrowth of DRG neurons of different chemical phenotypes which express or lack the InsR. Insulin, at a concentration of 10 nM, significantly increased total neurite length, the length of the longest neurite and the number of branch points of cultured DRG neurons as compared to neurons cultured in control medium or in the presence of 1 μM insulin. In both the control and the insulin exposed cultures, ∼43% of neurons displayed InsR-immunoreactivity. The proportions of transient receptor potential vanilloid type 1 receptor (TRPV1)-immunoreactive (IR), calcitonin gene-related peptide (CGRP)-IR and Bandeiraea simplicifolia isolectin B4 (IB4)-binding neurons amounted to ∼61%, ∼57%, and ∼31% of DRG neurons IR for the InsR. Of the IB4-positive population only neurons expressing the InsR were responsive to insulin. In contrast, TRPV1-IR nociceptive and CGRP-IR peptidergic neurons showed increased tendency for neurite outgrowth which was further enhanced by insulin. However, the responsiveness of DRG neurons expressing the InsR was superior to populations of DRG neurons which lack this receptor. The findings also revealed that besides the expression of the InsR, inherent properties of peptidergic, but not non-peptidergic nociceptive neurons may also significantly contribute to the mechanisms of neurite outgrowth of DRG neurons. These observations suggest distinct regenerative propensity for differing populations of DRG neurons which is significantly affected through insulin receptor signaling.
Collapse
Affiliation(s)
- Bence András Lázár
- Department of Psychiatry, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Department of Physiology, University of Szeged, Szeged, Hungary
| | - Gábor Jancsó
- Department of Physiology, University of Szeged, Szeged, Hungary
| | - Laura Pálvölgyi
- Department of Physiology, University of Szeged, Szeged, Hungary
| | - Ildikó Dobos
- Department of Physiology, University of Szeged, Szeged, Hungary
| | - István Nagy
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Péter Sántha
- Department of Physiology, University of Szeged, Szeged, Hungary
| |
Collapse
|
32
|
Duarte A, Santos M, Oliveira C, Moreira P. Brain insulin signalling, glucose metabolism and females' reproductive aging: A dangerous triad in Alzheimer's disease. Neuropharmacology 2018; 136:223-242. [PMID: 29471055 DOI: 10.1016/j.neuropharm.2018.01.044] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/22/2018] [Accepted: 01/29/2018] [Indexed: 12/12/2022]
|
33
|
Jin L, Lin L, Li GY, Liu S, Luo DJ, Feng Q, Sun DS, Wang W, Liu JJ, Wang Q, Ke D, Yang XF, Liu GP. Monosodium glutamate exposure during the neonatal period leads to cognitive deficits in adult Sprague-Dawley rats. Neurosci Lett 2018; 682:39-44. [PMID: 29885453 DOI: 10.1016/j.neulet.2018.06.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 05/20/2018] [Accepted: 06/05/2018] [Indexed: 02/05/2023]
Abstract
Epidemiological surveys show that 70-80% of patients with Alzheimer's disease (AD) have type 2 diabetes mellitus (T2DM) or show an abnormality of blood glucose levels. Therefore, an increasing number of evidence has suggested that diabetic hyperglycemia is tightly linked with the pathogenesis and progression of AD. In the present study, we replicated T2DM animal model via subcutaneous injection of newborn Sprague-Dawley (SD) rats with monosodium glutamate (MSG) during the neonatal period to investigate the effects and underlying mechanisms of hyperglycemia on cognitive ability. We found that neonatal MSG exposure induced hyperglycemia as well as Alzheimer-like learning and memory deficits with decreased dendritic spine density and hippocampal synaptic-related protein expression and increased phosphorylated tau levels in ∼3-month-old SD rats. Our results suggested that hyperglycemia probably causes cognitive impairment and Alzheimer-like neuropathological changes, which provide the experimental data connecting T2DM and AD.
Collapse
Affiliation(s)
- Li Jin
- Department of Pathophysiology, Henan Medical College, Zhengzhou 451191, China; Henan Medical Key Laboratory of Cerebrodegenerative Disease, Henan Medical College, Zhengzhou 451191, China.
| | - Li Lin
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan 430030, China; Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan 430030, China
| | - Guo-Yong Li
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan 430030, China; Department of Cardiology, West China Hospital, Sichuan University, 37 Guo Xue Xiang,Chengdu 610041, China
| | - Sha Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan 430030, China; Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Dan-Ju Luo
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qiong Feng
- Department of Pathology, Wuhan Children's Hospital, Wuhan, 430016, China
| | - Dong-Sheng Sun
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan 430030, China
| | - Wei Wang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan 430030, China
| | - Jian-Jun Liu
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, No. 8 Longyuan Road, Nanshan District, Shenzhen 518055, China
| | - Qun Wang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan 430030, China
| | - Dan Ke
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan 430030, China
| | - Xi-Fei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, No. 8 Longyuan Road, Nanshan District, Shenzhen 518055, China.
| | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan 430030, China; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, JS 226001, China.
| |
Collapse
|
34
|
Salgado-Puga K, Rodríguez-Colorado J, Prado-Alcalá RA, Peña-Ortega F. Subclinical Doses of ATP-Sensitive Potassium Channel Modulators Prevent Alterations in Memory and Synaptic Plasticity Induced by Amyloid-β. J Alzheimers Dis 2018; 57:205-226. [PMID: 28222502 DOI: 10.3233/jad-160543] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In addition to coupling cell metabolism and excitability, ATP-sensitive potassium channels (KATP) are involved in neural function and plasticity. Moreover, alterations in KATP activity and expression have been observed in Alzheimer's disease (AD) and during amyloid-β (Aβ)-induced pathology. Thus, we tested whether KATP modulators can influence Aβ-induced deleterious effects on memory, hippocampal network function, and plasticity. We found that treating animals with subclinical doses (those that did not change glycemia) of a KATP blocker (Tolbutamide) or a KATP opener (Diazoxide) differentially restrained Aβ-induced memory deficit, hippocampal network activity inhibition, and long-term synaptic plasticity unbalance (i.e., inhibition of LTP and promotion of LTD). We found that the protective effect of Tolbutamide against Aβ-induced memory deficit was strong and correlated with the reestablishment of synaptic plasticity balance, whereas Diazoxide treatment produced a mild protection against Aβ-induced memory deficit, which was not related to a complete reestablishment of synaptic plasticity balance. Interestingly, treatment with both KATP modulators renders the hippocampus resistant to Aβ-induced inhibition of hippocampal network activity. These findings indicate that KATP are involved in Aβ-induced pathology and they heighten the potential role of KATP modulation as a plausible therapeutic strategy against AD.
Collapse
Affiliation(s)
- Karla Salgado-Puga
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| | - Javier Rodríguez-Colorado
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| | - Roberto A Prado-Alcalá
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| |
Collapse
|
35
|
Kennedy G, Hardman RJ, Macpherson H, Scholey AB, Pipingas A. How Does Exercise Reduce the Rate of Age-Associated Cognitive Decline? A Review of Potential Mechanisms. J Alzheimers Dis 2018; 55:1-18. [PMID: 27636853 DOI: 10.3233/jad-160665] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The rate of age-associated cognitive decline varies considerably between individuals. It is important, both on a societal and individual level, to investigate factors that underlie these differences in order to identify those which might realistically slow cognitive decline. Physical activity is one such factor with substantial support in the literature. Regular exercise can positively influence cognitive ability, reduce the rate of cognitive aging, and even reduce the risk of Alzheimer's disease (AD) and other dementias. However, while there is substantial evidence in the extant literature for the effect of exercise on cognition, the processes that mediate this relationship are less clear. This review examines cardiovascular health, production of brain derived neurotrophic factor (BDNF), insulin sensitivity, stress, and inflammation as potential pathways, via which exercise may maintain or improve cognitive functioning, and may be particularly pertinent in the context of the aging brain. A greater understanding of these mechanisms and their potential relationships with exercise and cognition will be invaluable in providing biomarkers for investigating the efficacy of differing exercise regimes on cognitive outcomes.
Collapse
Affiliation(s)
- Greg Kennedy
- Centre for Human Psychopharmacology, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Roy J Hardman
- Centre for Human Psychopharmacology, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Helen Macpherson
- Centre for Human Psychopharmacology, Swinburne University of Technology, Melbourne, VIC, Australia.,Centre for Physical Activity and Nutrition Research, Deakin University, Melbourne, VIC, Australia
| | - Andrew B Scholey
- Centre for Human Psychopharmacology, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Andrew Pipingas
- Centre for Human Psychopharmacology, Swinburne University of Technology, Melbourne, VIC, Australia
| |
Collapse
|
36
|
Intranasal insulin treatment alleviates methamphetamine induced anxiety-like behavior and neuroinflammation. Neurosci Lett 2017; 660:122-129. [DOI: 10.1016/j.neulet.2017.09.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 09/11/2017] [Accepted: 09/12/2017] [Indexed: 01/03/2023]
|
37
|
Kullmann S, Heni M, Hallschmid M, Fritsche A, Preissl H, Häring HU. Brain Insulin Resistance at the Crossroads of Metabolic and Cognitive Disorders in Humans. Physiol Rev 2016; 96:1169-209. [PMID: 27489306 DOI: 10.1152/physrev.00032.2015] [Citation(s) in RCA: 361] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ever since the brain was identified as an insulin-sensitive organ, evidence has rapidly accumulated that insulin action in the brain produces multiple behavioral and metabolic effects, influencing eating behavior, peripheral metabolism, and cognition. Disturbances in brain insulin action can be observed in obesity and type 2 diabetes (T2D), as well as in aging and dementia. Decreases in insulin sensitivity of central nervous pathways, i.e., brain insulin resistance, may therefore constitute a joint pathological feature of metabolic and cognitive dysfunctions. Modern neuroimaging methods have provided new means of probing brain insulin action, revealing the influence of insulin on both global and regional brain function. In this review, we highlight recent findings on brain insulin action in humans and its impact on metabolism and cognition. Furthermore, we elaborate on the most prominent factors associated with brain insulin resistance, i.e., obesity, T2D, genes, maternal metabolism, normal aging, inflammation, and dementia, and on their roles regarding causes and consequences of brain insulin resistance. We also describe the beneficial effects of enhanced brain insulin signaling on human eating behavior and cognition and discuss potential applications in the treatment of metabolic and cognitive disorders.
Collapse
Affiliation(s)
- Stephanie Kullmann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Tübingen, Germany; Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany; Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; and Department of Pharmacy and Biochemistry, Faculty of Science, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Martin Heni
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Tübingen, Germany; Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany; Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; and Department of Pharmacy and Biochemistry, Faculty of Science, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Manfred Hallschmid
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Tübingen, Germany; Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany; Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; and Department of Pharmacy and Biochemistry, Faculty of Science, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Tübingen, Germany; Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany; Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; and Department of Pharmacy and Biochemistry, Faculty of Science, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Hubert Preissl
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Tübingen, Germany; Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany; Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; and Department of Pharmacy and Biochemistry, Faculty of Science, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Tübingen, Germany; Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany; Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; and Department of Pharmacy and Biochemistry, Faculty of Science, Eberhard Karls Universität Tübingen, Tübingen, Germany
| |
Collapse
|
38
|
Abstract
Recent results suggest that insulin is synthesised by a subpopulation of neurons in the cerebral cortex and neural progenitor cells of the hippocampus. Supplementing the slow supply of insulin to the brain by pancreatic beta cells, the insulin locally released by neurons provides a rapid means of regulating local microcircuits, effectively modulating synaptic transmission and on-demand energy homeostasis of neural networks. Modulation of insulin production by brain neurons via glucagon-like peptide 1 (GLP-1) agonists might be useful in counteracting diabetes, obesity and neurodegenerative diseases. Replacement of lost pancreatic beta cells by autologous transplantation of insulin-producing neural progenitor cells could be a viable therapy for diabetes.
Collapse
Affiliation(s)
- Éva A Csajbók
- Endocrine Unit, 1st Department of Internal Medicine, University of Szeged, Szeged, Hungary
| | - Gábor Tamás
- MTA-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy and Neuroscience, University of Szeged, Közép Fasor 52, Szeged, 6726, Hungary.
| |
Collapse
|
39
|
Abstract
There is well accepted evidence that the long-term outcome of schizophrenia is better in developing than in developed countries. Socio-cultural factors, which are as yet unidentified, have been postulated to explain this. There is also disputed evidence that schizophrenia was rare in indigenous populations and increased when they came into contact with Western or industrialised cultures, and that there was an increase in the rate of schizophrenia during the Industrial Revolution. These epidemiological and historical findings point to diet as a possible mediating factor in schizophrenia. Ecological studies have indicated that a worse outcome of schizophrenia is associated with higher consumption of saturated fat and sugar. It is also known that schizophrenic patients have increased insulin resistance and are at increased risk of developing diabetes and coronary heart disease. These findings suggest a new paradigm for our understanding of the causation and treatment of schizophrenia.
Collapse
|
40
|
Sestile CC, Maraschin JC, Rangel MP, Cuman RKN, Audi EA. Antidepressant-like Effect of Insulin in Streptozotocin-induced Type 2 Diabetes Mellitus Rats. Basic Clin Pharmacol Toxicol 2016; 119:243-8. [DOI: 10.1111/bcpt.12563] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/02/2016] [Indexed: 12/29/2022]
Affiliation(s)
- Caio C. Sestile
- Department of Pharmacology and Therapeutics; State University of Maringa; Maringa Brazil
| | - Jhonatan C. Maraschin
- Department of Pharmacology and Therapeutics; State University of Maringa; Maringa Brazil
| | - Marcel P. Rangel
- Department of Pharmacology and Therapeutics; State University of Maringa; Maringa Brazil
| | - Roberto K. N. Cuman
- Department of Pharmacology and Therapeutics; State University of Maringa; Maringa Brazil
| | - Elisabeth A. Audi
- Department of Pharmacology and Therapeutics; State University of Maringa; Maringa Brazil
| |
Collapse
|
41
|
Ma Y, Tang N, Thompson RC, Mobley BC, Clark SW, Sarkaria JN, Wang J. InsR/IGF1R Pathway Mediates Resistance to EGFR Inhibitors in Glioblastoma. Clin Cancer Res 2015; 22:1767-76. [PMID: 26561558 DOI: 10.1158/1078-0432.ccr-15-1677] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/23/2015] [Indexed: 01/09/2023]
Abstract
PURPOSE Aberrant activation of EGFR is a hallmark of glioblastoma. However, EGFR inhibitors exhibit at best modest efficacy in glioblastoma. This is in sharp contrast with the observations in EGFR-mutant lung cancer. We examined whether activation of functionally redundant receptor tyrosine kinases (RTKs) conferred resistance to EGFR inhibitors in glioblastoma. EXPERIMENTAL DESIGN We collected a panel of patient-derived glioblastoma xenograft (PDX) lines that maintained expression of wild-type or mutant EGFR in serial xenotransplantation and tissue cultures. Using this physiologically relevant platform, we tested the abilities of several RTK ligands to protect glioblastoma cells against an EGFR inhibitor, gefitinib. Based on the screening results, we further developed a combination therapy cotargeting EGFR and insulin receptor (InsR)/insulin-like growth factor 1 receptor (IGF1R). RESULTS Insulin and IGF1 induced significant protection against gefitinib in the majority of EGFR-dependent PDX lines with one exception that did not express InsR or IGF1R. Blockade of the InsR/IGF1R pathway synergistically improved sensitivity to gefitinib or dacomitinib. Gefitinib alone effectively attenuated EGFR activities and the downstream MEK/ERK pathway. However, repression of AKT and induction of apoptosis required concurrent inhibition of both EGFR and InsR/IGF1R. A combination of gefitinib and OSI-906, a dual InsR/IGF1R inhibitor, was more effective than either agent alone to treat subcutaneous glioblastoma xenograft tumors. CONCLUSIONS Our results suggest that activation of the InsR/IGF1R pathway confers resistance to EGFR inhibitors in EGFR-dependent glioblastoma through AKT regulation. Concurrent blockade of these two pathways holds promise to treat EGFR-dependent glioblastoma.
Collapse
Affiliation(s)
- Yufang Ma
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Nan Tang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Reid C Thompson
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Bret C Mobley
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Steven W Clark
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Jialiang Wang
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee. Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee. Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
42
|
Rosiglitazone improves learning and memory ability in rats with type 2 diabetes through the insulin signaling pathway. Am J Med Sci 2015; 350:121-8. [PMID: 25973687 DOI: 10.1097/maj.0000000000000499] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Diabetes mellitus (DM) is associated with moderate cognitive deficits and neurophysiologic and structural changes in the brain, a condition that is referred to as diabetic encephalopathy. This study was performed to investigate the effect of rosiglitazone (RSG) on learning and memory in rats with DM and elucidate possible mechanisms underlying this condition. Thirty-two male Sprague-Dawley rats were randomly divided into 4 groups: control (C, n = 8), DM (n = 8), RSG-administered control (C + RSG, n = 8) and RSG-administered DM groups (DM + RSG, n = 8). At 8 weeks after drug administration, Morris water maze was used to perform a training and probe trial to detect spatial learning and memory abilities. Western blot and immunohistochemistry were also used to detect changes in proteins involved in the insulin signal transduction pathway, such as the insulin receptor, insulin receptor substrate-1, protein kinase B, phosphorylated cAMP response element-binding protein and B-cell lymphoma 2, in the hippocampus of the rats. This study found that RSG could normalize the impaired insulin signal transduction in type 2 DM. The authors showed that RSG modulated the central insulin signaling axis.
Collapse
|
43
|
Streptozotocin Intracerebroventricular-Induced Neurotoxicity and Brain Insulin Resistance: a Therapeutic Intervention for Treatment of Sporadic Alzheimer's Disease (sAD)-Like Pathology. Mol Neurobiol 2015; 53:4548-62. [PMID: 26298663 DOI: 10.1007/s12035-015-9384-y] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 08/10/2015] [Indexed: 02/03/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is remarkably characterized by pathological hallmarks which include amyloid plaques, neurofibrillary tangles, neuronal loss, and progressive cognitive loss. Several well-known genetic mutations which are being used for the development of a transgenic model of AD lead to an early onset familial AD (fAD)-like condition. However, these settings are only reasons for a small percentage of the total AD cases. The large majorities of AD cases are considered as a sporadic in origin and are less influenced by a single mutation of a gene. The etiology of sporadic Alzheimer's disease (sAD) remains unclear, but numerous risk factors have been identified that increase the chance of developing AD. Among these risk factors are insulin desensitization/resistance state, oxidative stress, neuroinflammation, synapse dysfunction, tau hyperphosphorylation, and deposition of Aβ in the brain. Subsequently, these risk factors lead to development of sAD. However, the underlying molecular mechanism is not so clear. Streptozotocin (STZ) produces similar characteristic pathology of sAD such as altered glucose metabolism, insulin signaling, synaptic dysfunction, protein kinases such as protein kinase B/C, glycogen synthase-3β (GSK-3β) activation, tau hyperphosphorylation, Aβ deposition, and neuronal apoptosis. Further, STZ also leads to inhibition of Akt/PKB, insulin receptor (IR) signaling molecule, and insulin resistance in brain. These alterations mediated by STZ can be used to explore the underlying molecular and pathophysiological mechanism of AD (especially sAD) and their therapeutic intervention for drug development against AD pathology.
Collapse
|
44
|
Gong Y, Ma Y, Sinyuk M, Loganathan S, Thompson RC, Sarkaria JN, Chen W, Lathia JD, Mobley BC, Clark SW, Wang J. Insulin-mediated signaling promotes proliferation and survival of glioblastoma through Akt activation. Neuro Oncol 2015; 18:48-57. [PMID: 26136493 DOI: 10.1093/neuonc/nov096] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 05/07/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Metabolic complications such as obesity, hyperglycemia, and type 2 diabetes are associated with poor outcomes in patients with glioblastoma. To control peritumoral edema, use of chronic high-dose steroids in glioblastoma patients is common, which can result in de novo diabetic symptoms. These metabolic complications may affect tumors via profound mechanisms, including activation of insulin receptor (InsR) and the related insulin-like growth factor 1 receptor (IGF1R) in malignant cells. METHODS In the present study, we assessed expression of InsR in glioblastoma surgical specimens and glioblastoma response to insulin at physiologically relevant concentrations. We further determined whether genetic or pharmacological targeting of InsR affected oncogenic functions of glioblastoma in vitro and in vivo. RESULTS We showed that InsR was commonly expressed in glioblastoma surgical specimens and xenograft tumor lines, with mitogenic isoform-A predominating. Insulin at physiologically relevant concentrations promoted glioblastoma cell growth and survival, potentially via Akt activation. Depletion of InsR impaired cellular functions and repressed orthotopic tumor growth. The absence of InsR compromised downstream Akt activity, but yet stimulated IGF1R expression. Targeting both InsR and IGF1R with dual kinase inhibitors resulted in effective blockade of downstream signaling, loss of cell viability, and repression of xenograft tumor growth. CONCLUSIONS Taken together, our work suggests that glioblastoma is sensitive to the mitogenic functions of insulin, thus significant insulin exposure imposes risks to glioblastoma patients. Additionally, dual inhibition of InsR and IGF1R exhibits promise for treating glioblastoma.
Collapse
Affiliation(s)
- Yuanying Gong
- Department of Neurological Surgery (Y.G., Y.M., R.C.T., S.W.C., J.W.), Department of Molecular Physiology and Biophysics (W.C.), Department of Neurology (S.W.C.), Department of Pathology, Microbiology and Immunology (B.C.M.), and Department of Cancer Biology and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (J.W.); Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio (M.S., J.D.L.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Yufang Ma
- Department of Neurological Surgery (Y.G., Y.M., R.C.T., S.W.C., J.W.), Department of Molecular Physiology and Biophysics (W.C.), Department of Neurology (S.W.C.), Department of Pathology, Microbiology and Immunology (B.C.M.), and Department of Cancer Biology and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (J.W.); Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio (M.S., J.D.L.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Maksim Sinyuk
- Department of Neurological Surgery (Y.G., Y.M., R.C.T., S.W.C., J.W.), Department of Molecular Physiology and Biophysics (W.C.), Department of Neurology (S.W.C.), Department of Pathology, Microbiology and Immunology (B.C.M.), and Department of Cancer Biology and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (J.W.); Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio (M.S., J.D.L.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Sudan Loganathan
- Department of Neurological Surgery (Y.G., Y.M., R.C.T., S.W.C., J.W.), Department of Molecular Physiology and Biophysics (W.C.), Department of Neurology (S.W.C.), Department of Pathology, Microbiology and Immunology (B.C.M.), and Department of Cancer Biology and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (J.W.); Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio (M.S., J.D.L.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Reid C Thompson
- Department of Neurological Surgery (Y.G., Y.M., R.C.T., S.W.C., J.W.), Department of Molecular Physiology and Biophysics (W.C.), Department of Neurology (S.W.C.), Department of Pathology, Microbiology and Immunology (B.C.M.), and Department of Cancer Biology and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (J.W.); Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio (M.S., J.D.L.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Jann N Sarkaria
- Department of Neurological Surgery (Y.G., Y.M., R.C.T., S.W.C., J.W.), Department of Molecular Physiology and Biophysics (W.C.), Department of Neurology (S.W.C.), Department of Pathology, Microbiology and Immunology (B.C.M.), and Department of Cancer Biology and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (J.W.); Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio (M.S., J.D.L.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Wenbiao Chen
- Department of Neurological Surgery (Y.G., Y.M., R.C.T., S.W.C., J.W.), Department of Molecular Physiology and Biophysics (W.C.), Department of Neurology (S.W.C.), Department of Pathology, Microbiology and Immunology (B.C.M.), and Department of Cancer Biology and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (J.W.); Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio (M.S., J.D.L.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Justin D Lathia
- Department of Neurological Surgery (Y.G., Y.M., R.C.T., S.W.C., J.W.), Department of Molecular Physiology and Biophysics (W.C.), Department of Neurology (S.W.C.), Department of Pathology, Microbiology and Immunology (B.C.M.), and Department of Cancer Biology and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (J.W.); Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio (M.S., J.D.L.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Bret C Mobley
- Department of Neurological Surgery (Y.G., Y.M., R.C.T., S.W.C., J.W.), Department of Molecular Physiology and Biophysics (W.C.), Department of Neurology (S.W.C.), Department of Pathology, Microbiology and Immunology (B.C.M.), and Department of Cancer Biology and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (J.W.); Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio (M.S., J.D.L.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Stephen W Clark
- Department of Neurological Surgery (Y.G., Y.M., R.C.T., S.W.C., J.W.), Department of Molecular Physiology and Biophysics (W.C.), Department of Neurology (S.W.C.), Department of Pathology, Microbiology and Immunology (B.C.M.), and Department of Cancer Biology and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (J.W.); Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio (M.S., J.D.L.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Jialiang Wang
- Department of Neurological Surgery (Y.G., Y.M., R.C.T., S.W.C., J.W.), Department of Molecular Physiology and Biophysics (W.C.), Department of Neurology (S.W.C.), Department of Pathology, Microbiology and Immunology (B.C.M.), and Department of Cancer Biology and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (J.W.); Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio (M.S., J.D.L.); Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee (S.L.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| |
Collapse
|
45
|
Caravaggio F, Hahn M, Nakajima S, Gerretsen P, Remington G, Graff-Guerrero A. Reduced insulin-receptor mediated modulation of striatal dopamine release by basal insulin as a possible contributing factor to hyperdopaminergia in schizophrenia. Med Hypotheses 2015; 85:391-6. [PMID: 26118462 DOI: 10.1016/j.mehy.2015.06.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 05/22/2015] [Accepted: 06/17/2015] [Indexed: 12/23/2022]
Abstract
Schizophrenia is a severe and chronic neuropsychiatric disorder which affects 1% of the world population. Using the brain imaging technique positron emission tomography (PET) it has been demonstrated that persons with schizophrenia have greater dopamine transmission in the striatum compared to healthy controls. However, little progress has been made as to elucidating other biological mechanisms which may account for this hyperdopaminergic state in this disease. Studies in animals have demonstrated that insulin receptors are expressed on midbrain dopamine neurons, and that insulin from the periphery acts on these receptors to modify dopamine transmission in the striatum. This is pertinent given that several lines of evidence suggest that insulin receptor functioning may be abnormal in the brains of persons with schizophrenia. Post-mortem studies have shown that persons with schizophrenia have less than half the number of cortical insulin receptors compared to healthy persons. Moreover, these post-mortem findings are unlikely due to the effects of antipsychotic treatment; studies in cell lines and animals suggest antipsychotics enhance insulin receptor functioning. Further, hyperinsulinemia - even prior to antipsychotic use - seems to be related to less psychotic symptoms in patients with schizophrenia. Collectively, these data suggest that midbrain insulin receptor functioning may be abnormal in persons with schizophrenia, resulting in reduced insulin-mediated regulation of dopamine transmission in the striatum. Such a deficit may account for the hyperdopaminergic state observed in these patients and would help guide the development of novel treatment strategies. We hypothesize that, (i) insulin receptor expression and/or function is reduced in midbrain dopamine neurons in persons with schizophrenia, (ii) basal insulin should reduce dopaminergic transmission in the striatum via these receptors, and (iii) this modulation of dopaminergic transmission by basal insulin is reduced in the brains of persons with schizophrenia.
Collapse
Affiliation(s)
- Fernando Caravaggio
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario M5T 1R8, Canada; Institute of Medical Science, University of Toronto, 2374 Medical Sciences Building, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Margaret Hahn
- Institute of Medical Science, University of Toronto, 2374 Medical Sciences Building, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario M5T 1R8, Canada
| | - Shinichiro Nakajima
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario M5T 1R8, Canada
| | - Philip Gerretsen
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario M5T 1R8, Canada; Institute of Medical Science, University of Toronto, 2374 Medical Sciences Building, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario M5T 1R8, Canada
| | - Gary Remington
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario M5T 1R8, Canada; Institute of Medical Science, University of Toronto, 2374 Medical Sciences Building, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario M5T 1R8, Canada
| | - Ariel Graff-Guerrero
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario M5T 1R8, Canada; Institute of Medical Science, University of Toronto, 2374 Medical Sciences Building, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario M5T 1R8, Canada.
| |
Collapse
|
46
|
Abstract
Alzheimer's disease (AD) is a neurological disorder characterized by profound memory loss and progressive dementia. Accumulating evidence suggests that Type 2 diabetes mellitus, a metabolic disorder characterized by insulin resistance and glucose intolerance, significantly increases the risk for developing AD. Whereas amyloid-β (Aβ) deposition and neurofibrillary tangles are major histological hallmarks of AD, impairment of cerebral glucose metabolism precedes these pathological changes during the early stage of AD and likely triggers or exacerbates AD pathology. However, the mechanisms linking disturbed insulin signaling/glucose metabolism and AD pathogenesis remain unclear. The low-density lipoprotein receptor-related protein 1 (LRP1), a major apolipoprotein E receptor, plays critical roles in lipoprotein metabolism, synaptic maintenance, and clearance of Aβ in the brain. Here, we demonstrate that LRP1 interacts with the insulin receptor β in the brain and regulates insulin signaling and glucose uptake. LRP1 deficiency in neurons leads to impaired insulin signaling as well as reduced levels of glucose transporters GLUT3 and GLUT4. Consequently, glucose uptake is reduced. By using an in vivo microdialysis technique sampling brain glucose concentration in freely moving mice, we further show that LRP1 deficiency in conditional knock-out mice resulted in glucose intolerance in the brain. We also found that hyperglycemia suppresses LRP1 expression, which further exacerbates insulin resistance, glucose intolerance, and AD pathology. As loss of LRP1 expression is seen in AD brains, our study provides novel insights into insulin resistance in AD. Our work also establishes new targets that can be explored for AD prevention or therapy.
Collapse
|
47
|
Ribeiro IMR, Ferreira-Neto HC, Antunes VR. Subdiaphragmatic vagus nerve activity and hepatic venous glucose are differentially regulated by the central actions of insulin in Wistar and SHR. Physiol Rep 2015; 3:3/5/e12381. [PMID: 25948821 PMCID: PMC4463817 DOI: 10.14814/phy2.12381] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Glucose is the most important energy substrate for the maintenance of tissues function. The liver plays an essential role in the control of glucose production, since it is able to synthesize, store, and release glucose into the circulation under different situations. Hormones like insulin and catecholamines influence hepatic glucose production (HGP), but little is known about the role of the central actions of physiological doses of insulin in modulating HGP via the autonomic nervous system in nonanesthetized rats especially in SHR where we see a high degree of insulin resistance and metabolic dysfunction. Wistar and SHR received ICV injection of insulin (100 nU/μL) and hepatic venous glucose concentration (HVGC) was monitored for 30 min, as an indirect measure of HGP. At 10 min after insulin injection, HVGC decreased by 27% in Wistar rats, with a negligible change (3%) in SHR. Pretreatment with atropine totally blocked the reduction in HVGC, while pretreatment with propranolol and phentolamine induced a decrease of 8% in HVGC after ICV insulin injection in Wistar. Intracarotid infusion of insulin caused a significant increase in subdiaphragmatic vagus nerve (SVN) activity in Wistar (12 ± 2%), with negligible effects on the lumbar splanchnic sympathetic nerve (LSSN) activity (−6 ± 3%). No change was observed in SVN (−2 ± 2%) and LSSN activities (2 ± 3%) in SHR after ICA insulin infusion. Taken together, these results show, in nonanesthetized animals, the importance of the parasympathetic nervous system in controlling HVGC, and subdiaphragmatic nerve activity following central administration of insulin; a mechanism that is impaired in the SHR.
Collapse
Affiliation(s)
- Izabela Martina R Ribeiro
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo (USP), Sao Paulo, Brazil
| | - Hildebrando C Ferreira-Neto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo (USP), Sao Paulo, Brazil
| | - Vagner R Antunes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo (USP), Sao Paulo, Brazil
| |
Collapse
|
48
|
Lipid droplets hypertrophy: a crucial determining factor in insulin regulation by adipocytes. Sci Rep 2015; 5:8816. [PMID: 25743104 PMCID: PMC4649717 DOI: 10.1038/srep08816] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 01/27/2015] [Indexed: 01/14/2023] Open
Abstract
Lipid droplets (LDs) hypertrophy in adipocytes is the main cause of energy metabolic system dysfunction, obesity and its afflictions such as T2D. However, the role of adipocytes in linking energy metabolic disorders with insulin regulation is unknown in humans. Human adipocytes constitutively synthesize and secrete insulin, which is biologically functional. Insulin concentrations and release are fat mass- and LDs-dependent respectively. Fat reduction mediated by bariatric surgery repairs obesity-associated T2D. The expression of genes, like PCSK1 (proinsulin conversion enzyme), GCG (Glucagon), GPLD1, CD38 and NNAT, involved in insulin regulation/release were differentially expressed in pancreas and adipose tissue (AT). INS (insulin) and GCG expression reduced in human AT-T2D as compared to AT-control, but remained unchanged in pancreas in either state. Insulin levels (mRNA/protein) were higher in AT derived from prediabetes BB rats with destructed pancreatic β-cells and controls than pancreas derived from the same rats respectively. Insulin expression in 10 human primary cell types including adipocytes and macrophages is an evidence for extrapancreatic insulin-producing cells. The data suggest a crosstalk between AT and pancreas to fine-tune energy metabolic system or may minimize the metabolic damage during diabetes. This study opens new avenues towards T2D therapy with a great impact on public health.
Collapse
|
49
|
Specialized insulin is used for chemical warfare by fish-hunting cone snails. Proc Natl Acad Sci U S A 2015; 112:1743-8. [PMID: 25605914 DOI: 10.1073/pnas.1423857112] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
More than 100 species of venomous cone snails (genus Conus) are highly effective predators of fish. The vast majority of venom components identified and functionally characterized to date are neurotoxins specifically targeted to receptors, ion channels, and transporters in the nervous system of prey, predators, or competitors. Here we describe a venom component targeting energy metabolism, a radically different mechanism. Two fish-hunting cone snails, Conus geographus and Conus tulipa, have evolved specialized insulins that are expressed as major components of their venoms. These insulins are distinctive in having much greater similarity to fish insulins than to the molluscan hormone and are unique in that posttranslational modifications characteristic of conotoxins (hydroxyproline, γ-carboxyglutamate) are present. When injected into fish, the venom insulin elicits hypoglycemic shock, a condition characterized by dangerously low blood glucose. Our evidence suggests that insulin is specifically used as a weapon for prey capture by a subset of fish-hunting cone snails that use a net strategy to capture prey. Insulin appears to be a component of the nirvana cabal, a toxin combination in these venoms that is released into the water to disorient schools of small fish, making them easier to engulf with the snail's distended false mouth, which functions as a net. If an entire school of fish simultaneously experiences hypoglycemic shock, this should directly facilitate capture by the predatory snail.
Collapse
|
50
|
Akintola AA, van Heemst D. Insulin, aging, and the brain: mechanisms and implications. Front Endocrinol (Lausanne) 2015; 6:13. [PMID: 25705204 PMCID: PMC4319489 DOI: 10.3389/fendo.2015.00013] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 01/21/2015] [Indexed: 12/12/2022] Open
Abstract
There is now an impressive body of literature implicating insulin and insulin signaling in successful aging and longevity. New information from in vivo and in vitro studies concerning insulin and insulin receptors has extended our understanding of the physiological role of insulin in the brain. However, the relevance of these to aging and longevity remains to be elucidated. Here, we review advances in our understanding of the physiological role of insulin in the brain, how insulin gets into the brain, and its relevance to aging and longevity. Furthermore, we examine possible future therapeutic applications and implications of insulin in the context of available models of delayed and accelerated aging.
Collapse
Affiliation(s)
- Abimbola A. Akintola
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, Netherlands
- *Correspondence: Abimbola A. Akintola, Department of Gerontology and Geriatrics, Leiden University Medical Center, C7-124, Albinusdreef 2, Leiden 2333 ZA, Netherlands e-mail:
| | - Diana van Heemst
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|