Skip to main content
Top
Published in: Molecular Neurodegeneration 1/2017

Open Access 01-12-2017 | Research article

Intranasal insulin reverts central pathology and cognitive impairment in diabetic mother offspring

Authors: Juan Jose Ramos-Rodriguez, Daniel Sanchez-Sotano, Alberto Doblas-Marquez, Carmen Infante-Garcia, Simon Lubian-Lopez, Monica Garcia-Alloza

Published in: Molecular Neurodegeneration | Issue 1/2017

Login to get access

Abstract

Background

Adverse effects in diabetic mothers offspring (DMO) are a major concern of increasing incidence. Among these, chronic central complications in DMO remain poorly understood, and in extreme cases, diabetes can essentially function as a gestational brain insult. Nevertheless, therapeutic alternatives for DMO are limited.

Methods

Therefore, we have analyzed the central long-term complications in the offspring from CD1 diabetic mothers treated with streptozotozin, as well as the possible reversion of these alterations by insulin administration to neonates. Brain atrophy, neuronal morphology, tau phosphorylation, proliferation and neurogenesis were assessed in the short term (P7) and in the early adulthood (10 weeks) and cognitive function was also analyzed in the long-term.

Results

Central complications in DMO were still detected in the adulthood, including cortical and hippocampal thinning due to synaptic loss and neuronal simplification, increased tau hyperphosphorylation, and diminished cell proliferation and neurogenesis. Additionally, maternal diabetes increased the long-term susceptibility to spontaneous central bleeding, inflammation and cognition impairment in the offspring. On the other hand, intracerebroventricular insulin administration to neonates significantly reduced observed alterations. Moreover, non-invasive intranasal insulin reversed central atrophy and tau hyperphosphorylation, and rescued central proliferation and neurogenesis. Vascular damage, inflammation and cognitive alterations were also comparable to their counterparts born to nondiabetic mice, supporting the utility of this pathway to access the central nervous system.

Conclusions

Our data underlie the long-term effects of central complications in DMO. Moreover, observed improvement after insulin treatment opens the door to therapeutic alternatives for children who are exposed to poorly controlled gestational diabetes, and who may benefit from more individualized treatments.
Literature
1.
go back to reference Anna V, van der Ploeg HP, Cheung NW, Huxley RR, Bauman AE. Sociodemographic correlates of the increasing trend in prevalence of gestational diabetes mellitus in a large population of women between 1995 and 2005. Diabetes Care. 2008;31:2288–93.CrossRefPubMedPubMedCentral Anna V, van der Ploeg HP, Cheung NW, Huxley RR, Bauman AE. Sociodemographic correlates of the increasing trend in prevalence of gestational diabetes mellitus in a large population of women between 1995 and 2005. Diabetes Care. 2008;31:2288–93.CrossRefPubMedPubMedCentral
2.
go back to reference Metzger BE, Lowe LP, Dyer AR, Trimble ER, Chaovarindr U, Coustan DR, Hadden DR, McCance DR, Hod M, McIntyre HD, Oats JJ, Persson B, Rogers MS, Sacks DA. Hyperglycemia and adverse pregnancy outcomes. N Engl J Med. 2008;358:1991–2002.CrossRefPubMed Metzger BE, Lowe LP, Dyer AR, Trimble ER, Chaovarindr U, Coustan DR, Hadden DR, McCance DR, Hod M, McIntyre HD, Oats JJ, Persson B, Rogers MS, Sacks DA. Hyperglycemia and adverse pregnancy outcomes. N Engl J Med. 2008;358:1991–2002.CrossRefPubMed
4.
go back to reference Silverman BL, Metzger BE, Cho NH, Loeb CA. Impaired glucose tolerance in adolescent offspring of diabetic mothers. Relationship to fetal hyperinsulinism. Diabetes Care. 1995;18:611–7.CrossRefPubMed Silverman BL, Metzger BE, Cho NH, Loeb CA. Impaired glucose tolerance in adolescent offspring of diabetic mothers. Relationship to fetal hyperinsulinism. Diabetes Care. 1995;18:611–7.CrossRefPubMed
5.
go back to reference Piya MK, Harte AL, Sivakumar K, Tripathi G, Voyias PD, James S, Sabico S, Al-Daghri NM, Saravanan P, Barber TM, Kumar S, Vatish M, McTernan PG. The identification of irisin in human cerebrospinal fluid: influence of adiposity, metabolic markers, and gestational diabetes. Am J Physiol Endocrinol Metab. 2014;306:E512–8.CrossRefPubMed Piya MK, Harte AL, Sivakumar K, Tripathi G, Voyias PD, James S, Sabico S, Al-Daghri NM, Saravanan P, Barber TM, Kumar S, Vatish M, McTernan PG. The identification of irisin in human cerebrospinal fluid: influence of adiposity, metabolic markers, and gestational diabetes. Am J Physiol Endocrinol Metab. 2014;306:E512–8.CrossRefPubMed
6.
go back to reference Steculorum SM, Bouret SG. Maternal diabetes compromises the organization of hypothalamic feeding circuits and impairs leptin sensitivity in offspring. Endocrinology. 2011;152:4171–9.CrossRefPubMedPubMedCentral Steculorum SM, Bouret SG. Maternal diabetes compromises the organization of hypothalamic feeding circuits and impairs leptin sensitivity in offspring. Endocrinology. 2011;152:4171–9.CrossRefPubMedPubMedCentral
7.
go back to reference Dheen ST, Tay SS, Boran J, Ting LW, Kumar SD, Fu J, Ling EA. Recent studies on neural tube defects in embryos of diabetic pregnancy: an overview. Curr Med Chem. 2009;16:2345–54.CrossRefPubMed Dheen ST, Tay SS, Boran J, Ting LW, Kumar SD, Fu J, Ling EA. Recent studies on neural tube defects in embryos of diabetic pregnancy: an overview. Curr Med Chem. 2009;16:2345–54.CrossRefPubMed
8.
go back to reference Hami J, Vafaei-Nezhad S, Ghaemi K, Sadeghi A, Ivar G, Shojae F, Hosseini M. Stereological study of the effects of maternal diabetes on cerebellar cortex development in rat. Metab Brain Dis. 2016;31:643–52.CrossRefPubMed Hami J, Vafaei-Nezhad S, Ghaemi K, Sadeghi A, Ivar G, Shojae F, Hosseini M. Stereological study of the effects of maternal diabetes on cerebellar cortex development in rat. Metab Brain Dis. 2016;31:643–52.CrossRefPubMed
9.
go back to reference Perna R, Loughan AR, Le J, Tyson K. Gestational Diabetes: Long-Term Central Nervous System Developmental and Cognitive Sequelae. Appl Neuropsychol Child. 2015;4:217–20.CrossRefPubMed Perna R, Loughan AR, Le J, Tyson K. Gestational Diabetes: Long-Term Central Nervous System Developmental and Cognitive Sequelae. Appl Neuropsychol Child. 2015;4:217–20.CrossRefPubMed
10.
11.
go back to reference Aggarwal A, Khera A, Singh I, Sandhir R. S-nitrosoglutathione prevents blood–brain barrier disruption associated with increased matrix metalloproteinase-9 activity in experimental diabetes. J Neurochem. 2015;132:595–608.CrossRefPubMed Aggarwal A, Khera A, Singh I, Sandhir R. S-nitrosoglutathione prevents blood–brain barrier disruption associated with increased matrix metalloproteinase-9 activity in experimental diabetes. J Neurochem. 2015;132:595–608.CrossRefPubMed
12.
go back to reference Macedo RF, Furlan FC, Marshall PS, Michelotto JB, Gontijo JA. Effect of intracerebroventricularly injected insulin on urinary sodium excretion by cerebroventricular streptozotocin-treated rats. Braz J Med Biol Res. 2003;36:1193–9.CrossRefPubMed Macedo RF, Furlan FC, Marshall PS, Michelotto JB, Gontijo JA. Effect of intracerebroventricularly injected insulin on urinary sodium excretion by cerebroventricular streptozotocin-treated rats. Braz J Med Biol Res. 2003;36:1193–9.CrossRefPubMed
13.
go back to reference Koch C, Augustine RA, Steger J, Ganjam GK, Benzler J, Pracht C, Lowe C, Schwartz MW, Shepherd PR, Anderson GM, Grattan DR, Tups A. Leptin rapidly improves glucose homeostasis in obese mice by increasing hypothalamic insulin sensitivity. J Neurosci. 2010;30:16180–7.CrossRefPubMedPubMedCentral Koch C, Augustine RA, Steger J, Ganjam GK, Benzler J, Pracht C, Lowe C, Schwartz MW, Shepherd PR, Anderson GM, Grattan DR, Tups A. Leptin rapidly improves glucose homeostasis in obese mice by increasing hypothalamic insulin sensitivity. J Neurosci. 2010;30:16180–7.CrossRefPubMedPubMedCentral
14.
go back to reference Renner DB, Svitak AL, Gallus NJ, Ericson ME, Frey WH 2nd, Hanson LR. Intranasal delivery of insulin via the olfactory nerve pathway. J Pharm Pharmacol. 2012;64:1709–14.CrossRefPubMed Renner DB, Svitak AL, Gallus NJ, Ericson ME, Frey WH 2nd, Hanson LR. Intranasal delivery of insulin via the olfactory nerve pathway. J Pharm Pharmacol. 2012;64:1709–14.CrossRefPubMed
15.
go back to reference Marks DR, Tucker K, Cavallin MA, Mast TG, Fadool DA. Awake intranasal insulin delivery modifies protein complexes and alters memory, anxiety, and olfactory behaviors. J Neurosci. 2009;29:6734–51.CrossRefPubMedPubMedCentral Marks DR, Tucker K, Cavallin MA, Mast TG, Fadool DA. Awake intranasal insulin delivery modifies protein complexes and alters memory, anxiety, and olfactory behaviors. J Neurosci. 2009;29:6734–51.CrossRefPubMedPubMedCentral
16.
go back to reference Jimenez-Palomares M, Ramos-Rodriguez JJ, Lopez-Acosta JF, Pacheco-Herrero M, Lechuga-Sancho AM, Perdomo G, Garcia-Alloza M, Cozar-Castellano I. Increased Abeta production prompts the onset of glucose intolerance and insulin resistance. Am J Physiol Endocrinol Metab. 2012;302:E1373–80.CrossRefPubMed Jimenez-Palomares M, Ramos-Rodriguez JJ, Lopez-Acosta JF, Pacheco-Herrero M, Lechuga-Sancho AM, Perdomo G, Garcia-Alloza M, Cozar-Castellano I. Increased Abeta production prompts the onset of glucose intolerance and insulin resistance. Am J Physiol Endocrinol Metab. 2012;302:E1373–80.CrossRefPubMed
17.
go back to reference Ramos-Rodriguez JJ, Ortiz O, Jimenez-Palomares M, Kay KR, Berrocoso E, Murillo-Carretero MI, Perdomo G, Spires-Jones T, Cozar-Castellano I, Lechuga-Sancho AM, Garcia-Alloza M. Differential central pathology and cognitive impairment in pre-diabetic and diabetic mice. Psychoneuroendocrinology. 2013;38:2462–75.CrossRefPubMed Ramos-Rodriguez JJ, Ortiz O, Jimenez-Palomares M, Kay KR, Berrocoso E, Murillo-Carretero MI, Perdomo G, Spires-Jones T, Cozar-Castellano I, Lechuga-Sancho AM, Garcia-Alloza M. Differential central pathology and cognitive impairment in pre-diabetic and diabetic mice. Psychoneuroendocrinology. 2013;38:2462–75.CrossRefPubMed
18.
go back to reference Ramos-Rodriguez JJ, Molina-Gil S, Ortiz-Barajas O, Jimenez-Palomares M, Perdomo G, Cozar-Castellano I, Lechuga-Sancho AM, Garcia-Alloza M. Central proliferation and neurogenesis is impaired in type 2 diabetes and prediabetes animal models. PLoS One. 2014;9:e89229.CrossRefPubMedPubMedCentral Ramos-Rodriguez JJ, Molina-Gil S, Ortiz-Barajas O, Jimenez-Palomares M, Perdomo G, Cozar-Castellano I, Lechuga-Sancho AM, Garcia-Alloza M. Central proliferation and neurogenesis is impaired in type 2 diabetes and prediabetes animal models. PLoS One. 2014;9:e89229.CrossRefPubMedPubMedCentral
19.
go back to reference Ramos-Rodriguez JJ, Infante-Garcia C, Galindo-Gonzalez L, Garcia-Molina Y, Lechuga-Sancho A, Garcia-Alloza M. Increased Spontaneous Central Bleeding and Cognition Impairment in APP/PS1 Mice with Poorly Controlled Diabetes Mellitus. Mol Neurobiol. 2016;53:2685–97.CrossRefPubMed Ramos-Rodriguez JJ, Infante-Garcia C, Galindo-Gonzalez L, Garcia-Molina Y, Lechuga-Sancho A, Garcia-Alloza M. Increased Spontaneous Central Bleeding and Cognition Impairment in APP/PS1 Mice with Poorly Controlled Diabetes Mellitus. Mol Neurobiol. 2016;53:2685–97.CrossRefPubMed
20.
go back to reference Infante-Garcia C, Ramos-Rodriguez JJ, Galindo-Gonzalez L, Garcia-Alloza M. Long-term central pathology and cognitive impairment are exacerbated in a mixed model of Alzheimer's disease and type 2 diabetes. Psychoneuroendocrinology. 2015;65:15–25.CrossRefPubMed Infante-Garcia C, Ramos-Rodriguez JJ, Galindo-Gonzalez L, Garcia-Alloza M. Long-term central pathology and cognitive impairment are exacerbated in a mixed model of Alzheimer's disease and type 2 diabetes. Psychoneuroendocrinology. 2015;65:15–25.CrossRefPubMed
21.
go back to reference Ramos-Rodriguez JJ, Spires-Jones T, Pooler AM, Lechuga-Sancho AM, Bacskai BJ, Garcia-Alloza M. Progressive Neuronal Pathology and Synaptic Loss Induced by Prediabetes and Type 2 Diabetes in a Mouse Model of Alzheimer’s Disease. Mol Neurobiol. 2016: in press. Ramos-Rodriguez JJ, Spires-Jones T, Pooler AM, Lechuga-Sancho AM, Bacskai BJ, Garcia-Alloza M. Progressive Neuronal Pathology and Synaptic Loss Induced by Prediabetes and Type 2 Diabetes in a Mouse Model of Alzheimer’s Disease. Mol Neurobiol. 2016: in press.
22.
go back to reference Garcia-Alloza M, Borrelli LA, Rozkalne A, Hyman BT, Bacskai BJ. Curcumin labels amyloid pathology in vivo, disrupts existing plaques, and partially restores distorted neurites in an Alzheimer mouse model. J Neurochem. 2007;102:1095–104.CrossRefPubMed Garcia-Alloza M, Borrelli LA, Rozkalne A, Hyman BT, Bacskai BJ. Curcumin labels amyloid pathology in vivo, disrupts existing plaques, and partially restores distorted neurites in an Alzheimer mouse model. J Neurochem. 2007;102:1095–104.CrossRefPubMed
23.
go back to reference Ramos-Rodriguez JJ, Jimenez-Palomares M, Murillo-Carretero MI, Infante-Garcia C, Berrocoso E, Hernandez-Pacho F, Lechuga-Sancho AM, Cozar-Castellano I, Garcia-Alloza M. Central vascular disease and exacerbated pathology in a mixed model of type 2 diabetes and Alzheimer's disease. Psychoneuroendocrinology. 2015;62:69–79.CrossRefPubMed Ramos-Rodriguez JJ, Jimenez-Palomares M, Murillo-Carretero MI, Infante-Garcia C, Berrocoso E, Hernandez-Pacho F, Lechuga-Sancho AM, Cozar-Castellano I, Garcia-Alloza M. Central vascular disease and exacerbated pathology in a mixed model of type 2 diabetes and Alzheimer's disease. Psychoneuroendocrinology. 2015;62:69–79.CrossRefPubMed
25.
go back to reference Camprubi Robles M, Campoy C, Garcia Fernandez L, Lopez-Pedrosa JM, Rueda R, Martin MJ. Maternal Diabetes and Cognitive Performance in the Offspring: A Systematic Review and Meta-Analysis. PLoS One. 2015;10:e0142583.CrossRefPubMedPubMedCentral Camprubi Robles M, Campoy C, Garcia Fernandez L, Lopez-Pedrosa JM, Rueda R, Martin MJ. Maternal Diabetes and Cognitive Performance in the Offspring: A Systematic Review and Meta-Analysis. PLoS One. 2015;10:e0142583.CrossRefPubMedPubMedCentral
26.
go back to reference Hami J, Kerachian MA, Karimi R, Haghir H, Sadr-Nabavi A. Effects of streptozotocin-induced type 1 maternal diabetes on PI3K/AKT signaling pathway in the hippocampus of rat neonates. J Recept Signal Transduct Res. 2016;36:254–60.CrossRefPubMed Hami J, Kerachian MA, Karimi R, Haghir H, Sadr-Nabavi A. Effects of streptozotocin-induced type 1 maternal diabetes on PI3K/AKT signaling pathway in the hippocampus of rat neonates. J Recept Signal Transduct Res. 2016;36:254–60.CrossRefPubMed
27.
go back to reference Sandeep MS, Nandini CD. Brain heparan sulphate proteoglycans are altered in developing foetus when exposed to in-utero hyperglycaemia. Metab Brain Dis. 2017. Sandeep MS, Nandini CD. Brain heparan sulphate proteoglycans are altered in developing foetus when exposed to in-utero hyperglycaemia. Metab Brain Dis. 2017.
28.
go back to reference Blondeau B, Joly B, Perret C, Prince S, Bruneval P, Lelievre-Pegorier M, Fassot C, Duong Van Huyen JP. Exposure in utero to maternal diabetes leads to glucose intolerance and high blood pressure with no major effects on lipid metabolism. Diabetes Metab. 2011;37:245–51.CrossRefPubMed Blondeau B, Joly B, Perret C, Prince S, Bruneval P, Lelievre-Pegorier M, Fassot C, Duong Van Huyen JP. Exposure in utero to maternal diabetes leads to glucose intolerance and high blood pressure with no major effects on lipid metabolism. Diabetes Metab. 2011;37:245–51.CrossRefPubMed
29.
go back to reference Heni M, Wagner R, Kullmann S, Veit R, Mat Husin H, Linder K, Benkendorff C, Peter A, Stefan N, Haring HU, Preissl H, Fritsche A. Central insulin administration improves whole-body insulin sensitivity via hypothalamus and parasympathetic outputs in men. Diabetes. 2014;63:4083–8.CrossRefPubMed Heni M, Wagner R, Kullmann S, Veit R, Mat Husin H, Linder K, Benkendorff C, Peter A, Stefan N, Haring HU, Preissl H, Fritsche A. Central insulin administration improves whole-body insulin sensitivity via hypothalamus and parasympathetic outputs in men. Diabetes. 2014;63:4083–8.CrossRefPubMed
30.
go back to reference Ott V, Benedict C, Schultes B, Born J, Hallschmid M. Intranasal administration of insulin to the brain impacts cognitive function and peripheral metabolism. Diabetes Obes Metab. 2012;14:214–21.CrossRefPubMed Ott V, Benedict C, Schultes B, Born J, Hallschmid M. Intranasal administration of insulin to the brain impacts cognitive function and peripheral metabolism. Diabetes Obes Metab. 2012;14:214–21.CrossRefPubMed
31.
go back to reference Chandna AR, Kuhlmann N, Bryce CA, Greba Q, Campanucci VA, Howland JG. Chronic maternal hyperglycemia induced during mid-pregnancy in rats increases RAGE expression, augments hippocampal excitability, and alters behavior of the offspring. Neuroscience. 2015;303:241–60.CrossRefPubMed Chandna AR, Kuhlmann N, Bryce CA, Greba Q, Campanucci VA, Howland JG. Chronic maternal hyperglycemia induced during mid-pregnancy in rats increases RAGE expression, augments hippocampal excitability, and alters behavior of the offspring. Neuroscience. 2015;303:241–60.CrossRefPubMed
32.
go back to reference Holscher C. First clinical data of the neuroprotective effects of nasal insulin application in patients with Alzheimer’s disease. Alzheimers Dement. 2014;10:S33–7.CrossRefPubMed Holscher C. First clinical data of the neuroprotective effects of nasal insulin application in patients with Alzheimer’s disease. Alzheimers Dement. 2014;10:S33–7.CrossRefPubMed
33.
go back to reference Novak V, Milberg W, Hao Y, Munshi M, Novak P, Galica A, Manor B, Roberson P, Craft S, Abduljalil A. Enhancement of vasoreactivity and cognition by intranasal insulin in type 2 diabetes. Diabetes Care. 2014;37:751–9.CrossRefPubMedPubMedCentral Novak V, Milberg W, Hao Y, Munshi M, Novak P, Galica A, Manor B, Roberson P, Craft S, Abduljalil A. Enhancement of vasoreactivity and cognition by intranasal insulin in type 2 diabetes. Diabetes Care. 2014;37:751–9.CrossRefPubMedPubMedCentral
34.
go back to reference Francis GJ, Martinez JA, Liu WQ, Xu K, Ayer A, Fine J, Tuor UI, Glazner G, Hanson LR, Frey WH 2nd, Toth C. Intranasal insulin prevents cognitive decline, cerebral atrophy and white matter changes in murine type I diabetic encephalopathy. Brain. 2008;131:3311–34.CrossRefPubMed Francis GJ, Martinez JA, Liu WQ, Xu K, Ayer A, Fine J, Tuor UI, Glazner G, Hanson LR, Frey WH 2nd, Toth C. Intranasal insulin prevents cognitive decline, cerebral atrophy and white matter changes in murine type I diabetic encephalopathy. Brain. 2008;131:3311–34.CrossRefPubMed
35.
go back to reference Hami J, Sadr-Nabavi A, Sankian M, Balali-Mood M, Haghir H. The effects of maternal diabetes on expression of insulin-like growth factor-1 and insulin receptors in male developing rat hippocampus. Brain Struct Funct. 2013;218:73–84.CrossRefPubMed Hami J, Sadr-Nabavi A, Sankian M, Balali-Mood M, Haghir H. The effects of maternal diabetes on expression of insulin-like growth factor-1 and insulin receptors in male developing rat hippocampus. Brain Struct Funct. 2013;218:73–84.CrossRefPubMed
36.
go back to reference Stern EA, Bacskai BJ, Hickey GA, Attenello FJ, Lombardo JA, Hyman BT. Cortical synaptic integration in vivo is disrupted by amyloid-beta plaques. J Neurosci. 2004;24(19):4535-40. Stern EA, Bacskai BJ, Hickey GA, Attenello FJ, Lombardo JA, Hyman BT. Cortical synaptic integration in vivo is disrupted by amyloid-beta plaques. J Neurosci. 2004;24(19):4535-40.
37.
go back to reference Vafaei-Nezhad S, Hami J, Sadeghi A, Ghaemi K, Hosseini M, Abedini MR, Haghir H. The impacts of diabetes in pregnancy on hippocampal synaptogenesis in rat neonates. Neuroscience. 2016;318:122–33.CrossRefPubMed Vafaei-Nezhad S, Hami J, Sadeghi A, Ghaemi K, Hosseini M, Abedini MR, Haghir H. The impacts of diabetes in pregnancy on hippocampal synaptogenesis in rat neonates. Neuroscience. 2016;318:122–33.CrossRefPubMed
38.
go back to reference Vogt MC, Paeger L, Hess S, Steculorum SM, Awazawa M, Hampel B, Neupert S, Nicholls HT, Mauer J, Hausen AC, Predel R, Kloppenburg P, Horvath TL, Bruning JC. Neonatal insulin action impairs hypothalamic neurocircuit formation in response to maternal high-fat feeding. Cell. 2014;156:495–509.CrossRefPubMedPubMedCentral Vogt MC, Paeger L, Hess S, Steculorum SM, Awazawa M, Hampel B, Neupert S, Nicholls HT, Mauer J, Hausen AC, Predel R, Kloppenburg P, Horvath TL, Bruning JC. Neonatal insulin action impairs hypothalamic neurocircuit formation in response to maternal high-fat feeding. Cell. 2014;156:495–509.CrossRefPubMedPubMedCentral
39.
go back to reference Liu J, Speder P, Brand AH. Control of brain development and homeostasis by local and systemic insulin signalling. Diabetes Obes Metab. 2014;16(Suppl 1):16–20.CrossRefPubMed Liu J, Speder P, Brand AH. Control of brain development and homeostasis by local and systemic insulin signalling. Diabetes Obes Metab. 2014;16(Suppl 1):16–20.CrossRefPubMed
40.
go back to reference Kruse MS, Barutta J, Vega MC, Coirini H. Down regulation of the proliferation and apoptotic pathways in the embryonic brain of diabetic rats. Cell Mol Neurobiol. 2012;32:1031–7.CrossRefPubMed Kruse MS, Barutta J, Vega MC, Coirini H. Down regulation of the proliferation and apoptotic pathways in the embryonic brain of diabetic rats. Cell Mol Neurobiol. 2012;32:1031–7.CrossRefPubMed
41.
go back to reference Bell GA, Fadool DA. Awake, long-term intranasal insulin treatment does not affect object memory, odor discrimination, or reversal learning in mice. Physiol Behav. 2017;174:104–13.CrossRefPubMed Bell GA, Fadool DA. Awake, long-term intranasal insulin treatment does not affect object memory, odor discrimination, or reversal learning in mice. Physiol Behav. 2017;174:104–13.CrossRefPubMed
42.
go back to reference Kim B, Backus C, Oh S, Hayes JM, Feldman EL. Increased tau phosphorylation and cleavage in mouse models of type 1 and type 2 diabetes. Endocrinology. 2009;150:5294–301.CrossRefPubMedPubMedCentral Kim B, Backus C, Oh S, Hayes JM, Feldman EL. Increased tau phosphorylation and cleavage in mouse models of type 1 and type 2 diabetes. Endocrinology. 2009;150:5294–301.CrossRefPubMedPubMedCentral
43.
go back to reference Martin SA, Jameson CH, Allan SM, Lawrence CB. Maternal high-fat diet worsens memory deficits in the triple-transgenic (3xTgAD) mouse model of Alzheimer's disease. PLoS One. 2014;9:e99226.CrossRefPubMedPubMedCentral Martin SA, Jameson CH, Allan SM, Lawrence CB. Maternal high-fat diet worsens memory deficits in the triple-transgenic (3xTgAD) mouse model of Alzheimer's disease. PLoS One. 2014;9:e99226.CrossRefPubMedPubMedCentral
44.
go back to reference Hankey GJ, Anderson NE, Ting RD, Veillard AS, Romo M, Wosik M, Sullivan DR, O'Connell RL, Hunt D, Keech AC. Rates and predictors of risk of stroke and its subtypes in diabetes: a prospective observational study. J Neurol Neurosurg Psychiatry. 2013;84:281–7.CrossRefPubMed Hankey GJ, Anderson NE, Ting RD, Veillard AS, Romo M, Wosik M, Sullivan DR, O'Connell RL, Hunt D, Keech AC. Rates and predictors of risk of stroke and its subtypes in diabetes: a prospective observational study. J Neurol Neurosurg Psychiatry. 2013;84:281–7.CrossRefPubMed
45.
go back to reference Zabrodina VV, Shreder ED, Shreder OV, Durnev AD, Seredenin SB. Impaired prenatal development and glycemic status in the offspring of rats with experimental streptozotocin-induced diabetes and their correction with afobazole. Bull Exp Biol Med. 2014;158:16–20.CrossRefPubMed Zabrodina VV, Shreder ED, Shreder OV, Durnev AD, Seredenin SB. Impaired prenatal development and glycemic status in the offspring of rats with experimental streptozotocin-induced diabetes and their correction with afobazole. Bull Exp Biol Med. 2014;158:16–20.CrossRefPubMed
46.
go back to reference Wen Y, Yang S, Liu R, Brun-Zinkernagel AM, Koulen P, Simpkins JW. Transient cerebral ischemia induces aberrant neuronal cell cycle re-entry and Alzheimer's disease-like tauopathy in female rats. J Biol Chem. 2004;279:22684–92.CrossRefPubMed Wen Y, Yang S, Liu R, Brun-Zinkernagel AM, Koulen P, Simpkins JW. Transient cerebral ischemia induces aberrant neuronal cell cycle re-entry and Alzheimer's disease-like tauopathy in female rats. J Biol Chem. 2004;279:22684–92.CrossRefPubMed
47.
go back to reference Zhang Q, Gao T, Luo Y, Chen X, Gao G, Gao X, Zhou Y, Dai J. Transient focal cerebral ischemia/reperfusion induces early and chronic axonal changes in rats: its importance for the risk of Alzheimer’s disease. PLoS One. 2012;7:e33722.CrossRefPubMedPubMedCentral Zhang Q, Gao T, Luo Y, Chen X, Gao G, Gao X, Zhou Y, Dai J. Transient focal cerebral ischemia/reperfusion induces early and chronic axonal changes in rats: its importance for the risk of Alzheimer’s disease. PLoS One. 2012;7:e33722.CrossRefPubMedPubMedCentral
48.
go back to reference Grayson BE, Levasseur PR, Williams SM, Smith MS, Marks DL, Grove KL. Changes in melanocortin expression and inflammatory pathways in fetal offspring of nonhuman primates fed a high-fat diet. Endocrinology. 2010;151:1622–32.CrossRefPubMedPubMedCentral Grayson BE, Levasseur PR, Williams SM, Smith MS, Marks DL, Grove KL. Changes in melanocortin expression and inflammatory pathways in fetal offspring of nonhuman primates fed a high-fat diet. Endocrinology. 2010;151:1622–32.CrossRefPubMedPubMedCentral
Metadata
Title
Intranasal insulin reverts central pathology and cognitive impairment in diabetic mother offspring
Authors
Juan Jose Ramos-Rodriguez
Daniel Sanchez-Sotano
Alberto Doblas-Marquez
Carmen Infante-Garcia
Simon Lubian-Lopez
Monica Garcia-Alloza
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Molecular Neurodegeneration / Issue 1/2017
Electronic ISSN: 1750-1326
DOI
https://doi.org/10.1186/s13024-017-0198-4

Other articles of this Issue 1/2017

Molecular Neurodegeneration 1/2017 Go to the issue