1
|
Dey J, Chandra S, Gupta J, Tripathi PP. Hippocampal neurodegeneration induces transient endogenous regeneration and long-term exhaustion of the neurogenic niche. J Cell Physiol 2024; 239:e31249. [PMID: 38501376 DOI: 10.1002/jcp.31249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 02/25/2024] [Accepted: 02/28/2024] [Indexed: 03/20/2024]
Abstract
The hippocampal dentate gyrus, responds to diverse pathological stimuli through neurogenesis. This phenomenon, observed following brain injury or neurodegeneration, is postulated to contribute to neuronal repair and functional recovery, thereby presenting an avenue for endogenous neuronal restoration. This study investigated the extent of regenerative response in hippocampal neurogenesis by leveraging the well-established kainic acid-induced status epilepticus model in vivo. In our study, we observed the activation and proliferation of neuronal progenitors or neural stem cell (NSC) and their subsequent migration to the injury sites following the seizure. At the injury sites, new neurons (Tuj1+BrdU+ and NeuN+BrdU+) have been generated indicating regenerative and reparative roles of the progenitor cells. We further detected whether this transient neurogenic burst, which might be a response towards an attempt to repair the brain, is associated with persistent long-term exhaustion of the dentate progenitor cells and impairment of adult neurogenesis marked by downregulation of Ki67, HoPX, and Sox2 with BrdU+ cell in the later part of life. Our studies suggest that the adult brain has the constitutive endogenous regenerative potential for brain repair to restore the damaged neurons, meanwhile, in the long term, it accelerates the depletion of the finite NSC pool in the hippocampal neurogenic niche by changing its proliferative and neurogenic capacity. A thorough understanding of the impact of modulating adult neurogenesis will eventually be required to design novel therapeutics to stimulate or assist brain repair while simultaneously preventing the adverse effects of early robust neurogenesis on the proliferative potential of endogenous neuronal progenitors.
Collapse
Affiliation(s)
- Jhilik Dey
- Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sreyashi Chandra
- Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Jalaj Gupta
- Stem Cell Research Centre, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| | - Prem Prakash Tripathi
- Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
2
|
Vargas-Soria M, García-Alloza M, Corraliza-Gómez M. Effects of diabetes on microglial physiology: a systematic review of in vitro, preclinical and clinical studies. J Neuroinflammation 2023; 20:57. [PMID: 36869375 PMCID: PMC9983227 DOI: 10.1186/s12974-023-02740-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/16/2023] [Indexed: 03/05/2023] Open
Abstract
Diabetes mellitus is a heterogeneous chronic metabolic disorder characterized by the presence of hyperglycemia, commonly preceded by a prediabetic state. The excess of blood glucose can damage multiple organs, including the brain. In fact, cognitive decline and dementia are increasingly being recognized as important comorbidities of diabetes. Despite the largely consistent link between diabetes and dementia, the underlying causes of neurodegeneration in diabetic patients remain to be elucidated. A common factor for almost all neurological disorders is neuroinflammation, a complex inflammatory process in the central nervous system for the most part orchestrated by microglial cells, the main representatives of the immune system in the brain. In this context, our research question aimed to understand how diabetes affects brain and/or retinal microglia physiology. We conducted a systematic search in PubMed and Web of Science to identify research items addressing the effects of diabetes on microglial phenotypic modulation, including critical neuroinflammatory mediators and their pathways. The literature search yielded 1327 records, including 18 patents. Based on the title and abstracts, 830 papers were screened from which 250 primary research papers met the eligibility criteria (original research articles with patients or with a strict diabetes model without comorbidities, that included direct data about microglia in the brain or retina), and 17 additional research papers were included through forward and backward citations, resulting in a total of 267 primary research articles included in the scoping systematic review. We reviewed all primary publications investigating the effects of diabetes and/or its main pathophysiological traits on microglia, including in vitro studies, preclinical models of diabetes and clinical studies on diabetic patients. Although a strict classification of microglia remains elusive given their capacity to adapt to the environment and their morphological, ultrastructural and molecular dynamism, diabetes modulates microglial phenotypic states, triggering specific responses that include upregulation of activity markers (such as Iba1, CD11b, CD68, MHC-II and F4/80), morphological shift to amoeboid shape, secretion of a wide variety of cytokines and chemokines, metabolic reprogramming and generalized increase of oxidative stress. Pathways commonly activated by diabetes-related conditions include NF-κB, NLRP3 inflammasome, fractalkine/CX3CR1, MAPKs, AGEs/RAGE and Akt/mTOR. Altogether, the detailed portrait of complex interactions between diabetes and microglia physiology presented here can be regarded as an important starting point for future research focused on the microglia-metabolism interface.
Collapse
Affiliation(s)
- María Vargas-Soria
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain.,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain
| | - Mónica García-Alloza
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain.,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain
| | - Miriam Corraliza-Gómez
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain. .,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain.
| |
Collapse
|
3
|
Marissal-Arvy N, Moisan MP. Diabetes and associated cognitive disorders: Role of the Hypothalamic-Pituitary Adrenal axis. Metabol Open 2022; 15:100202. [PMID: 35958117 PMCID: PMC9357829 DOI: 10.1016/j.metop.2022.100202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 11/12/2022] Open
Abstract
Both diabetes types, types 1 and 2, are associated with cognitive impairments. Each period of life is concerned, and this is an increasing public health problem. Animal models have been developed to investigate the biological actors involved in such impairments. Many levels of the brain function (structure, volume, neurogenesis, neurotransmission, behavior) are involved. In this review, we detailed the part potentially played by the Hypothalamic-Pituitary Adrenal axis in these dysfunctions. Notably, regulating glucocorticoid levels, their receptors and their bioavailability appear to be relevant for future research studies, and treatment development.
Collapse
Affiliation(s)
- Nathalie Marissal-Arvy
- INRAE, Laboratoire de Nutrition et Neurobiologie Intégrée, UMR 1286, UFR de Pharmacie, 146 Rue Léo Saignat, 33076, Bordeaux Cedex, France
| | - Marie-Pierre Moisan
- University of Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33000, Bordeaux, France
| |
Collapse
|
4
|
Steriade C, Titulaer MJ, Vezzani A, Sander JW, Thijs RD. The association between systemic autoimmune disorders and epilepsy and its clinical implications. Brain 2021; 144:372-390. [PMID: 33221878 DOI: 10.1093/brain/awaa362] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 08/03/2020] [Accepted: 08/13/2020] [Indexed: 12/12/2022] Open
Abstract
Systemic autoimmune disorders occur more frequently in patients with epilepsy than in the general population, suggesting shared disease mechanisms. The risk of epilepsy is elevated across the spectrum of systemic autoimmune disorders but is highest in systemic lupus erythematosus and type 1 diabetes mellitus. Vascular and metabolic factors are the most important mediators between systemic autoimmune disorders and epilepsy. Systemic immune dysfunction can also affect neuronal excitability, not only through innate immune activation and blood-brain barrier dysfunction in most epilepsies but also adaptive immunity in autoimmune encephalitis. The presence of systemic autoimmune disorders in subjects with acute seizures warrants evaluation for infectious, vascular, toxic and metabolic causes of acute symptomatic seizures, but clinical signs of autoimmune encephalitis should not be missed. Immunosuppressive medications may have antiseizure properties and trigger certain drug interactions with antiseizure treatments. A better understanding of mechanisms underlying the co-existence of epilepsy and systemic autoimmune disorders is needed to guide new antiseizure and anti-epileptogenic treatments. This review aims to summarize the epidemiological evidence for systemic autoimmune disorders as comorbidities of epilepsy, explore potential immune and non-immune mechanisms, and provide practical implications on diagnostic and therapeutic approach to epilepsy in those with comorbid systemic autoimmune disorders.
Collapse
Affiliation(s)
- Claude Steriade
- Department of Neurology, New York University School of Medicine, New York, NY, USA
| | - Maarten J Titulaer
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Annamaria Vezzani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Josemir W Sander
- NIHR University College London Hospitals Biomedical Research Centre, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK.,Chalfont Centre for Epilepsy, Chalfont St Peter SL9 0RJ, Bucks, UK.,Stichting Epilepsie Instellingen Nederland - (SEIN), Heemstede, The Netherlands
| | - Roland D Thijs
- NIHR University College London Hospitals Biomedical Research Centre, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK.,Stichting Epilepsie Instellingen Nederland - (SEIN), Heemstede, The Netherlands.,Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
5
|
Zinc in the Brain: Friend or Foe? Int J Mol Sci 2020; 21:ijms21238941. [PMID: 33255662 PMCID: PMC7728061 DOI: 10.3390/ijms21238941] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/22/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022] Open
Abstract
Zinc is a trace metal ion in the central nervous system that plays important biological roles, such as in catalysis, structure, and regulation. It contributes to antioxidant function and the proper functioning of the immune system. In view of these characteristics of zinc, it plays an important role in neurophysiology, which leads to cell growth and cell proliferation. However, after brain disease, excessively released and accumulated zinc ions cause neurotoxic damage to postsynaptic neurons. On the other hand, zinc deficiency induces degeneration and cognitive decline disorders, such as increased neuronal death and decreased learning and memory. Given the importance of balance in this context, zinc is a biological component that plays an important physiological role in the central nervous system, but a pathophysiological role in major neurological disorders. In this review, we focus on the multiple roles of zinc in the brain.
Collapse
|
6
|
Evaluation of neurotoxicity and hepatotoxicity effects of acute and sub-acute oral administration of unripe ackee ( Blighia sapida) fruit extract. Toxicol Rep 2019; 6:656-665. [PMID: 31338305 PMCID: PMC6626071 DOI: 10.1016/j.toxrep.2019.06.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 12/22/2022] Open
Abstract
Acute oral dose of 2000 mg/kg of unripe B. sapida fruit extract (BSE) was toxic to mice. Repeated treatment with BSE impaired locomotor function, memory performance and shortened seizure latency in mice. Repeated treatment with BSE significantly up-regulate acetylcholinesterase enzyme activity in mice. Repeated treatment with BSE elevates oxidative stress in the brain and liver of mice. Repeated treatment with BSE showed histopathological evidences of toxicity in mice brain and liver.
Ackee (Blighia sapida) is a commonly eaten fruit that is indigenous to West Africa and Jamaica. Ackee poisoning in young children have been reported in parts of Nigeria due to consumption of the unripe fruits. This study was designed to identify potential mechanisms of acute and sub-acute toxicity of unripe B. sapida fruit extract (BSE). Acute toxic effect was investigated in mice of either sex administered BSE 2000 mg/kg. The sub-acute toxicity effects were investigated in mice of either sex that received 28 days repeated administration of BSE (100 and 500 mg/kg, p.o.). Locomotor activity and memory performance were measured as well as seizure vulnerability in PTZ-induced model. Liver enzymes were assessed in the serum. Acetylcholinesterase, oxidative stress parameters and histopathological changes were assessed in the brain and liver tissues. Signs and symptoms of toxicity such as urination, tremor, depressed locomotion and death were observed in acute toxicity test. Sub-acute dosing caused significant impairment in locomotor activity and memory performance in mice. Seizure threshold was shortened in BSE-treated compared to control mice. Brain acetylcholinesterase activity was significantly increased. Alkaline phosphatase (ALP) was significantly elevated in mice that received BSE (500 mg/kg). Furthermore, BSE caused significant increase in oxidative stress expressed in nitrite, malondialdehyde, reduced glutathione and catalase in the brain and liver tissues. Histological staining revealed neuronal damage of brain hippocampus and hepatocellular swelling and necrosis. Blighia sapida unripe fruit extract increased susceptibility to seizure and impaired locomotor and memory function. The biochemical and histopathological findings revealed potential toxicity mechanisms related to neurotoxicity and hepatotoxicity.
Collapse
|
7
|
Nistor M, Schmidt M, Graul I, Rakers F, Schiffner R. A Systematic Review of Neuroprotective Strategies in the Management of Hypoglycemia. Int J Mol Sci 2019; 20:ijms20030550. [PMID: 30696060 PMCID: PMC6386855 DOI: 10.3390/ijms20030550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 01/18/2019] [Accepted: 01/24/2019] [Indexed: 02/07/2023] Open
Abstract
Severe hypogylcemia has been found to induce cerebral damage. While a number of illnesses can lead to hypoglycemic episodes, antidiabetic medications prescribed for glycemic control are a common cause. Considering the rising prevalence of diabetes mellitus in the population, we investigated neuroprotective strategies during hypoglycemia in the form of a systematic review in adherence to the PRISMA statement. A review protocol was registered in the PROSPERO database. A systematic literature search of PubMed, Web of Science, and CENTRAL was performed in September 2018. Based on a predefined inclusion protocol, results were screened and evaluated by two researchers. Both animal experiments and human studies were included, and their risk of bias was assessed with SYRCLE’s and the Cochrane risk of bias tools, respectively. Of a total of 16,230 results, 145 were assessed in full-text form: 27 articles adhered to the inclusion criteria and were qualitatively analyzed. The retrieved neuroprotective strategies could be categorized into three subsets: (1) Energy substitution, (2) hypoglycemia unawareness, and (3) other neuroprotective strategies. While on a study level, the individual results appeared promising, more research is required to investigate not only specific neuroprotective strategies against hypoglycemic cerebral damage, but also its underlying pathophysiological mechanisms.
Collapse
Affiliation(s)
- Marius Nistor
- Department of Neurology, Jena University Hospital - Friedrich Schiller University, Jena 07747, Germany.
| | - Martin Schmidt
- Institute for Biochemistry II, Jena University Hospital - Friedrich Schiller University, Jena 07743, Germany.
| | - Isabel Graul
- Orthopedic Department, Jena University Hospital - Friedrich Schiller University, Campus Eisenberg, Klosterlausnitzer Straße 81, Eisenberg 07607, Germany.
| | - Florian Rakers
- Department of Neurology, Jena University Hospital - Friedrich Schiller University, Jena 07747, Germany.
| | - René Schiffner
- Department of Neurology, Jena University Hospital - Friedrich Schiller University, Jena 07747, Germany.
- Orthopedic Department, Jena University Hospital - Friedrich Schiller University, Campus Eisenberg, Klosterlausnitzer Straße 81, Eisenberg 07607, Germany.
| |
Collapse
|
8
|
Zinc Promotes Adipose-Derived Mesenchymal Stem Cell Proliferation and Differentiation towards a Neuronal Fate. Stem Cells Int 2018; 2018:5736535. [PMID: 29765417 PMCID: PMC5932442 DOI: 10.1155/2018/5736535] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/08/2018] [Accepted: 03/18/2018] [Indexed: 01/28/2023] Open
Abstract
Zinc is an essential element required for cell division, migration, and proliferation. Under zinc-deficient conditions, proliferation and differentiation of neural progenitors are significantly impaired. Adipose-derived mesenchymal stem cells (AD-MSCs) are multipotent stem cells that can differentiate into neurons. The aim of this study was to evaluate the effect of zinc on AD-MSC proliferation and differentiation. We initially examined the effect of zinc on stem cell proliferation at the undifferentiated stage. AD-MSCs showed high proliferation rates on day 6 in 30 μM and 100 μM of ZnCl2. Zinc chelation inhibited AD-MSC proliferation via downregulation of ERK1/2 activity. We then assessed whether zinc was involved in cell migration and neurite outgrowth during differentiation. After three days of neuronal differentiation, TUJ-1-positive cells were observed, implying that AD-MSCs had differentiated into early neuron or neuron-like cells. Neurite outgrowth was increased in the zinc-treated group, while the CaEDTA-treated group showed diminished, shrunken neurites. Furthermore, we showed that zinc promoted neurite outgrowth via the inactivation of RhoA and led to the induction of neuronal gene expression (MAP2 and nestin) in differentiated stem cells. Taken together, zinc promoted AD-MSC proliferation and affected neuronal differentiation, mainly by increasing neurite outgrowth.
Collapse
|
9
|
Administration of placenta-derived mesenchymal stem cells counteracts a delayed anergic state following a transient induction of endogenous neurogenesis activity after global cerebral ischemia. Brain Res 2018; 1689:63-74. [PMID: 29625115 DOI: 10.1016/j.brainres.2018.03.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/25/2018] [Accepted: 03/28/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Global cerebral ischemia (GCI) is a major obstacle for cardiac arrest survival. Recent studies have suggested the possibility of mesenchymal stem cell (MSC) as a novel therapeutic option for GCI, but these results were limited to the neuroprotective effects of MSCs. Therefore, we aimed to investigate specific characteristics of neurogenesis after transient GCI, and to assess the effect of MSC on these characteristics. METHODS Adult male Sprague-Dawley rats were subjected to 7 min of transient GCI and randomized into 7 groups: baseline, MSC, and control administered groups, to be analyzed at 2, 3, and 4 weeks after GCI, respectively. The same interventions were repeated for sham operated animals. Rats were euthanized at the designated time after GCI. RESULTS A comparison of GCI and sham groups without MSC treatment, showed that the counts of bromodeoxyuridine (BrdU)- and doublecortin (DCX)-positive cells were significantly increased in the GCI group at 1 week after insult, but the trend was reversed at 3 weeks after insult. The counts of BrdU-, Ki67- and DCX-positive cells and the intensity of zinc translocator 3 (ZnT3) were all significantly higher in the MSC-treated group than those in the control group at 3 weeks after GCI. The count of NeuN-positive cells in the hippocampus was significantly increased in the MSC group at 4 weeks after GCI. CONCLUSIONS GCI induces transient neurogenesis, followed by an anergic state. MSC may counteract this anergy of neurogenesis and result in an increase in intact neurons in later stages.
Collapse
|
10
|
Administration of Zinc plus Cyclo-(His-Pro) Increases Hippocampal Neurogenesis in Rats during the Early Phase of Streptozotocin-Induced Diabetes. Int J Mol Sci 2017; 18:ijms18010073. [PMID: 28045430 PMCID: PMC5297708 DOI: 10.3390/ijms18010073] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 12/22/2016] [Accepted: 12/26/2016] [Indexed: 12/21/2022] Open
Abstract
The effects of zinc supplementation on hippocampal neurogenesis in diabetes mellitus have not been studied. Herein, we investigated the effects of zinc plus cyclo-(His-Pro) (ZC) on neurogenesis occurring in the subgranular zone of dentate gyrus after streptozotocin (STZ)-induced diabetes. ZC (27 mg/kg) was administered by gavage once daily for one or six weeks from the third day after the STZ injection, and histological evaluation was performed at 10 (early phase) or 45 (late phase) days after STZ injection. We found that the proliferation of progenitor cells in STZ-induced diabetic rats showed an increase in the early phase. Additionally, ZC treatment remarkably increased the number of neural progenitor cells (NPCs) and immature neurons in the early phase of STZ-induced diabetic rats. Furthermore, ZC treatment showed increased survival rate of newly generated cells but no difference in the level of neurogenesis in the late phase of STZ-induced diabetic rats. The present study demonstrates that zinc supplementation by ZC increases both NPCs proliferation and neuroblast production at the early phase of diabetes. Thus, this study suggests that zinc supplemented with a histidine/proline complex may have beneficial effects on neurogenesis in patients experiencing the early phase of Type 1 diabetes.
Collapse
|
11
|
Choi BY, Kim IY, Kim JH, Lee BE, Lee SH, Kho AR, Sohn M, Suh SW. Zinc plus cyclo-(His-Pro) promotes hippocampal neurogenesis in rats. Neuroscience 2016; 339:634-643. [DOI: 10.1016/j.neuroscience.2016.10.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 09/23/2016] [Accepted: 10/12/2016] [Indexed: 10/20/2022]
|
12
|
Mansouri S, Lietzau G, Lundberg M, Nathanson D, Nyström T, Patrone C. Pituitary Adenlylate Cyclase Activating Peptide Protects Adult Neural Stem Cells from a Hypoglycaemic milieu. PLoS One 2016; 11:e0156867. [PMID: 27305000 PMCID: PMC4909203 DOI: 10.1371/journal.pone.0156867] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 05/19/2016] [Indexed: 12/25/2022] Open
Abstract
Hypoglycaemia is a common side-effect of glucose-lowering therapies for type-2 diabetic patients, which may cause cognitive/neurological impairment. Although the effects of hypoglycaemia in the brain have been extensively studied in neurons, how hypoglycaemia impacts the viability of adult neural stem cells (NSCs) has been poorly investigated. In addition, the cellular and molecular mechanisms of how hypoglycaemia regulates NSCs survival have not been characterized. Recent work others and us have shown that the pituitary adenylate cyclase-activating polypeptide (PACAP) and the glucagon-like peptide-1 receptor (GLP-1R) agonist Exendin-4 stimulate NSCs survival against glucolipoapoptosis. The aim of this study was to establish an in vitro system where to study the effects of hypoglycaemia on NSC survival. Furthermore, we determine the potential role of PACAP and Exendin-4 in counteracting the effect of hypoglycaemia. A hypoglycaemic in vitro milieu was mimicked by exposing subventricular zone-derived NSC to low levels of glucose. Moreover, we studied the potential involvement of apoptosis and endoplasmic reticulum stress by quantifying protein levels of Bcl-2, cleaved caspase-3 and mRNA levels of CHOP. We show that PACAP via PAC-1 receptor and PKA activation counteracts impaired NSC viability induced by hypoglycaemia. The protective effect induced by PACAP correlated with endoplasmic reticulum stress, Exendin-4 was ineffective. The results show that hypoglycaemia decreases NSC viability and that this effect can be substantially counteracted by PACAP via PAC-1 receptor activation. The data supports a potential therapeutic role of PAC-1 receptor agonists for the treatment of neurological complications, based on neurogenesis impairment by hypoglycaemia.
Collapse
Affiliation(s)
- Shiva Mansouri
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| | - Grazyna Lietzau
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mathias Lundberg
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - David Nathanson
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Nyström
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Cesare Patrone
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
13
|
Bolaños L, Matute E, Ramírez-Dueñas MDL, Zarabozo D. Neuropsychological Impairment in School-Aged Children Born to Mothers With Gestational Diabetes. J Child Neurol 2015; 30:1616-24. [PMID: 25814475 DOI: 10.1177/0883073815575574] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 02/08/2015] [Indexed: 11/15/2022]
Abstract
The aim of this study was to determine whether school-aged children born to mothers with gestational diabetes show delays in their neuropsychological development. Several key neuropsychological characteristics of 32 children aged 7 to 9 years born to mothers with gestational diabetes were examined by comparing their performance on cognitive tasks to that of 28 children aged 8 to 10 years whose mothers had glucose levels within normal limits during pregnancy. The gestational diabetes group showed low performance on graphic, spatial, and bimanual skills and a higher presence of soft neurologic signs. Lower scores for general intellectual level and the working memory index were also evident. Our results suggest that gestational diabetes is associated with mild cognitive impairment.
Collapse
Affiliation(s)
- Lourdes Bolaños
- Instituto de Neurociencias, Universidad de Guadalajara, Guadalajara, Mexico
| | - Esmeralda Matute
- Instituto de Neurociencias, Universidad de Guadalajara, Guadalajara, Mexico
| | | | - Daniel Zarabozo
- Instituto de Neurociencias, Universidad de Guadalajara, Guadalajara, Mexico
| |
Collapse
|
14
|
Bachor TP, Marquioni-Ramella MD, Suburo AM. Sitagliptin protects proliferation of neural progenitor cells in diabetic mice. Metab Brain Dis 2015; 30:885-93. [PMID: 25694236 DOI: 10.1007/s11011-015-9656-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/03/2015] [Indexed: 12/14/2022]
Abstract
Sitagliptin (SIT) is a dipeptidyl peptidase-4 (DPP-4) inhibitor that enhances the effects of incretin hormones, such as Glucose-dependent Insulinotropic Peptide (also known as Gastric Inhibitory Polypeptide, GIP) and Glucagon-Like Peptide 1 (GLP-1). We have now evaluated the effect of SIT on proliferation of neural progenitors in diabetic mice. A condition resembling the non-obese type 2 diabetes mellitus (D2) was achieved by a combination of streptozotocin and nicotinamide (NA-STZ), whereas a type 1-like disease (D1) was provoked by STZ without NA. Non-diabetic mice received vehicle injections. Cell proliferation was estimated by bromodeoxyuridine (BrdU) incorporation in two different regions of the subventricular zone (SVZ), the largest reserve of neural stem cells in the adult brain. SIT treatment did not modify the high fasting blood glucose (BG) levels and intraperitoneal glucose tolerance test (IPGTT) of D1 mice. By contrast, in D2 mice, SIT treatment significantly reduced BG and IPGTT. Both D1 and D2 mice showed a substantial reduction of BrdU labeling in the SVZ. Remarkably, SIT treatment improved BrdU labeling in both conditions. Our findings suggest that SIT would protect proliferation of neural progenitor cells even in the presence of non-controlled diabetic alterations.
Collapse
Affiliation(s)
- Tomás P Bachor
- Medicina Celular y Molecular, Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, B1629AHJ, Argentina
| | | | | |
Collapse
|
15
|
Mauras N, Mazaika P, Buckingham B, Weinzimer S, White NH, Tsalikian E, Hershey T, Cato A, Cheng P, Kollman C, Beck RW, Ruedy K, Aye T, Fox L, Arbelaez AM, Wilson D, Tansey M, Tamborlane W, Peng D, Marzelli M, Winer KK, Reiss AL. Longitudinal assessment of neuroanatomical and cognitive differences in young children with type 1 diabetes: association with hyperglycemia. Diabetes 2015; 64:1770-9. [PMID: 25488901 PMCID: PMC4407847 DOI: 10.2337/db14-1445] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 12/05/2014] [Indexed: 12/11/2022]
Abstract
Significant regional differences in gray and white matter volume and subtle cognitive differences between young diabetic and nondiabetic children have been observed. Here, we assessed whether these differences change over time and the relation with dysglycemia. Children ages 4 to <10 years with (n = 144) and without (n = 72) type 1 diabetes (T1D) had high-resolution structural MRI and comprehensive neurocognitive tests at baseline and 18 months and continuous glucose monitoring and HbA1c performed quarterly for 18 months. There were no differences in cognitive and executive function scores between groups at 18 months. However, children with diabetes had slower total gray and white matter growth than control subjects. Gray matter regions (left precuneus, right temporal, frontal, and parietal lobes and right medial-frontal cortex) showed lesser growth in diabetes, as did white matter areas (splenium of the corpus callosum, bilateral superior-parietal lobe, bilateral anterior forceps, and inferior-frontal fasciculus). These changes were associated with higher cumulative hyperglycemia and glucose variability but not with hypoglycemia. Young children with T1D have significant differences in total and regional gray and white matter growth in brain regions involved in complex sensorimotor processing and cognition compared with age-matched control subjects over 18 months, suggesting that chronic hyperglycemia may be detrimental to the developing brain.
Collapse
Affiliation(s)
- Nelly Mauras
- Division of Endocrinology, Diabetes & Metabolism, Nemours Children's Clinic, Jacksonville, FL
| | - Paul Mazaika
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA
| | - Bruce Buckingham
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Stuart Weinzimer
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT
| | - Neil H White
- Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Eva Tsalikian
- Pediatric Endocrinology, University of Iowa, Iowa City, IA
| | - Tamara Hershey
- Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO Department of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Allison Cato
- Division of Neurology, Nemours Children's Clinic, Jacksonville, FL
| | | | | | - Roy W Beck
- Jaeb Center for Health Research, Tampa, FL
| | | | - Tandy Aye
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Larry Fox
- Division of Endocrinology, Diabetes & Metabolism, Nemours Children's Clinic, Jacksonville, FL
| | - Ana Maria Arbelaez
- Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Darrell Wilson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Michael Tansey
- Pediatric Endocrinology, University of Iowa, Iowa City, IA
| | - William Tamborlane
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT
| | - Daniel Peng
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA
| | - Matthew Marzelli
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA Department of Bioengineering, Stanford University School of Medicine, Stanford, CA
| | - Karen K Winer
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Allan L Reiss
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA Department of Pediatrics, Stanford University School of Medicine, Stanford, CA Department of Radiology, Stanford University School of Medicine, Stanford, CA
| | | |
Collapse
|
16
|
Tolcos M, Markwick R, O'Dowd R, Martin V, Turnley A, Rees S. Intrauterine Growth Restriction: Effects on Neural Precursor Cell Proliferation and Angiogenesis in the Foetal Subventricular Zone. Dev Neurosci 2015; 37:453-63. [PMID: 25720426 DOI: 10.1159/000371344] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/03/2014] [Indexed: 11/19/2022] Open
Abstract
Exposure to adverse prenatal factors can result in abnormal brain development, contributing to the aetiology of several neurological disorders. Intrauterine insults could occur during neurogenesis and gliogenesis, disrupting these events. Here we investigate the effects of chronic placental insufficiency (CPI) on cell proliferation and the microenvironment in the subventricular zone (SVZ). At 30 days of gestation (DG; term ∼67 DG), CPI was induced in pregnant guinea pigs via unilateral uterine artery ligation to produce growth-restricted (GR) foetuses (n = 7); controls (n = 6) were from the unoperated horn. At 60 DG, foetal brains were stained immunohistochemically to identify proliferating cells (Ki67), immature neurons (polysialylated neuronal cell adhesion molecule), astrocytes (glial fibrillary acidic protein), microglia (ionised calcium-binding adaptor molecule-1, Iba-1) and the microvasculature (von Willebrand factor) in the SVZ. There was no overall difference (p > 0.05) in the total number of Ki67-immunoreactive (IR) cells, the percentage of SVZ occupied by blood vessels or the density of Iba-1-IR microglia in control versus GR foetuses. However, regression analysis across both groups revealed that both the number of Ki67-IR cells and the percentage of SVZ occupied by blood vessels in the ventral SVZ were negatively correlated (p < 0.05) with brain weight. Furthermore, in the SVZ (dorsal and ventral) the density of blood vessels positively correlated (p < 0.05) with the number of Ki67-IR cells. Double-labelling immunofluorescence suggested that the majority of proliferating cells were likely to be neural precursor cells. Thus, we have demonstrated an association between angiogenesis and neurogenesis in the foetal neurogenic niche and have identified a window of opportunity for the administration of trophic support to enhance a neuroregenerative response.
Collapse
Affiliation(s)
- Mary Tolcos
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Vic., Australia
| | | | | | | | | | | |
Collapse
|
17
|
Choi BY, Kim JH, Kim HJ, Lee BE, Kim IY, Sohn M, Suh SW. Zinc chelation reduces traumatic brain injury-induced neurogenesis in the subgranular zone of the hippocampal dentate gyrus. J Trace Elem Med Biol 2014; 28:474-81. [PMID: 25200616 DOI: 10.1016/j.jtemb.2014.07.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Numerous studies have demonstrated that traumatic brain injury (TBI) increases hippocampal neurogenesis in the rodent brain. However, the mechanisms underlying increased neurogenesis after TBI remain unknown. Continuous neurogenesis occurs in the subgranular zone (SGZ) of the hippocampal dentate gyrus (DG) in the adult brain. The mechanism that maintains active neurogenesis in the hippocampal area is not known. A high level of vesicular zinc is localized in the presynaptic terminals of the SGZ (mossy fiber). The mossy fiber of dentate granular cells contains high levels of chelatable zinc in their terminal vesicles, which can be released into the extracellular space during neuronal activity. Previously, our lab presented findings indicating that a possible correlation may exist between synaptic zinc localization and high rates of neurogenesis in this area after hypoglycemia or epilepsy. Using a weight drop animal model to mimic human TBI, we tested our hypothesis that zinc plays a key role in modulating hippocampal neurogenesis after TBI. Thus, we injected a zinc chelator, clioquinol (CQ, 30mg/kg), into the intraperitoneal space to reduce brain zinc availability twice per day for 1 week. Neuronal death was evaluated with Fluoro Jade-B and NeuN staining to determine whether CQ has neuroprotective effects after TBI. The number of degenerating neurons (FJB (+)) and live neurons (NeuN (+)) was similar in vehicle and in CQ-treated rats at 1 week after TBI. Neurogenesis was evaluated using BrdU, Ki67 and doublecortin (DCX) immunostaining 1 week after TBI. The number of BrdU, Ki67 and DCX positive cell was increased after TBI. However, the number of BrdU, Ki67 and DCX positive cells was significantly decreased by CQ treatment. The present study shows that zinc chelation did not prevent neurodegeneration but did reduce TBI-induced progenitor cell proliferation and neurogenesis. Therefore, this study suggests that zinc has an essential role for modulating hippocampal neurogenesis after TBI.
Collapse
Affiliation(s)
- Bo Young Choi
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Republic of Korea
| | - Jin Hee Kim
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Republic of Korea
| | - Hyun Jung Kim
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Republic of Korea
| | - Bo Eun Lee
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Republic of Korea
| | - In Yeol Kim
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Republic of Korea
| | - Min Sohn
- Inha University, Department of Nursing, Incheon, Republic of Korea
| | - Sang Won Suh
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Republic of Korea.
| |
Collapse
|
18
|
Kim YH, Sung YH, Lee HH, Ko IG, Kim SE, Shin MS, Kim BK. Postnatal treadmill exercise alleviates short-term memory impairment by enhancing cell proliferation and suppressing apoptosis in the hippocampus of rat pups born to diabetic rats. J Exerc Rehabil 2014; 10:209-17. [PMID: 25210695 PMCID: PMC4157927 DOI: 10.12965/jer.140145] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 08/13/2014] [Indexed: 12/14/2022] Open
Abstract
During pregnancy, diabetes mellitus exerts detrimental effects on the development of the fetus, especially the central nervous system. In the current study, we evaluated the effects of postnatal treadmill exercise on short-term memory in relation with cell proliferation and apoptosis in the hippocampus of rat pups born to streptozotocin (STZ)-induced diabetic maternal rats. Adult female rats were mated with male rats for 24 h. Two weeks after mating, the pregnant female rats were divided into two groups: control group and STZ injection group. The pregnant rats in the STZ injection group were administered 40 mg/kg of STZ intraperitoneally. After birth, the rat pups were divided into the following four groups: control group, control with postnatal exercise group, maternal STZ-injection group, and maternal STZ-injection with postnatal exercise group. The rat pups in the postnatal exercise groups were made to run on a treadmill for 30 min once a day, 5 times per week for 2 weeks beginning 4 weeks after birth. The rat pups born to diabetic rats were shown to have short-term memory impairment with suppressed cell proliferation and increased apoptosis in the hippocampal dentate gyrus. Postnatal treadmill exercise alleviated short-term memory impairment by increased cell proliferation and suppressed apoptosis in the rat pups born to diabetic rats. These findings indicate that postnatal treadmill exercise may be used as a valuable strategy to ameliorate neurodevelopmental problems in children born to diabetics.
Collapse
Affiliation(s)
- Young Hoon Kim
- Department of Urology, Gachon University Gil Medical Center, Gachon University School of Medicine, Incheon, Korea
| | - Yun-Hee Sung
- Department of Physical Therapy, College of Natural Science, Kyungnam University, Changwon, Korea
| | - Hee-Hyuk Lee
- Department of Sports Science, College of Life Science and Nano Technology, Hannam University, Daejeon, Korea
| | - Il-Gyu Ko
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Sung-Eun Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Mal-Soon Shin
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Bo-Kyun Kim
- KBS Institute of the Sports, Arts and Science, Seoul, Korea
| |
Collapse
|
19
|
Kim DY, Rhee I, Paik J. Metabolic circuits in neural stem cells. Cell Mol Life Sci 2014; 71:4221-41. [PMID: 25037158 DOI: 10.1007/s00018-014-1686-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 06/25/2014] [Accepted: 07/14/2014] [Indexed: 12/15/2022]
Abstract
Metabolic activity indicative of cellular demand is emerging as a key player in cell fate decision. Numerous studies have demonstrated that diverse metabolic pathways have a critical role in the control of the proliferation, differentiation and quiescence of stem cells. The identification of neural stem/progenitor cells (NSPCs) and the characterization of their development and fate decision process have provided insight into the regenerative potential of the adult brain. As a result, the potential of NSPCs in cell replacement therapies for neurological diseases is rapidly growing. The aim of this review is to discuss the recent findings on the crosstalk among key regulators of NSPC development and the metabolic regulation crucial for the function and cell fate decisions of NSPCs. Fundamental understanding of the metabolic circuits in NSPCs may help to provide novel approaches for reactivating neurogenesis to treat degenerative brain conditions and cognitive decline.
Collapse
Affiliation(s)
- Do-Yeon Kim
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York City, NY, 10065, USA
| | | | | |
Collapse
|
20
|
Abstract
Hypoglycemia occurs in diabetic patients as a consequence of treatment with hypoglycemic agents, in insulinoma patients as a result of excessive insulin production, and in infants as a result of abnormal regulation of metabolism. Profound hypoglycemia can cause structural and functional disturbances in both the central (CNS) and the peripheral nervous system (PNS). The brain is damaged by a short and severe episode of hypoglycemia, whereas PNS pathology appears after a mild and prolonged episode. In the CNS, damaged mitochondria, elevated intracellular Ca2(+) level, released cytochrome c to the cytosol, extensive production of superoxide, increased caspase-3 activity, release of aspartate and glutamate from presynaptic terminals, and altered biosynthetic machinery can lead to neuronal cell death in the brain. Considering the PNS, chronic hypoglycemia is associated with delayed motor and sensory conduction velocities in peripheral nerves. With respect to pathology, hypoglycemic neuropathy in the PNS is characterized by Wallerian-like axonal degeneration that starts at the nerve terminal and progresses to a more proximal part of the axon, and motor axons to the muscles may be more severely damaged than sensory axons. Since excitatory neurotransmitters primarily involve the neuron in the CNS, this "dying back" pattern of axonal damage in the PNS may involve mechanisms other than excitotoxicity.
Collapse
Affiliation(s)
- Simin Mohseni
- Department of Clinical and Experimental Medicine, Division of Cell Biology, Faculty of Health Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
21
|
Mansouri S, Barde S, Ortsäter H, Eweida M, Darsalia V, Langel U, Sjöholm A, Hökfelt T, Patrone C. GalR3 activation promotes adult neural stem cell survival in response to a diabetic milieu. J Neurochem 2013; 127:209-20. [PMID: 23927369 DOI: 10.1111/jnc.12396] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 07/26/2013] [Accepted: 08/01/2013] [Indexed: 12/20/2022]
Abstract
Type 2 diabetes impairs adult neurogenesis which could play a role in the CNS complications of this serious disease. The goal of this study was to determine the potential role of galanin in protecting adult neural stem cells (NSCs) from glucolipotoxicity and to analyze whether apoptosis and the unfolded protein response were involved in the galanin-mediated effect. We also studied the regulation of galanin and its receptor subtypes under diabetes in NSCs in vitro and in the subventricular zone (SVZ) in vivo. The viability of mouse SVZ-derived NSCs and the involvement of apoptosis (Bcl-2, cleaved caspase-3) and unfolded protein response [C/EBP homologous protein (CHOP) Glucose-regulated protein 78/immunoglobulin heavy-chain binding protein (GRP78/BiP), spliced X-box binding protein 1 (XBP1), c-Jun N-terminal kinases (JNK) phosphorylation] were assessed in the presence of glucolipotoxic conditions after 24 h. The effect of diabetes on the regulation of galanin and its receptor subtypes was assessed on NSCs in vitro and in SVZ tissues isolated from normal and type 2 diabetes ob/ob mice. We show increased NSC viability following galanin receptor (GalR)3 activation. This protective effect correlated with decreased apoptosis and CHOP levels. We also report how galanin and its receptors are regulated by diabetes in vitro and in vivo. This study shows GalR3-mediated neuroprotection, supporting a potential future therapeutic development, based on GalR3 activation, for the treatment of brain disorders.
Collapse
Affiliation(s)
- Shiva Mansouri
- Karolinska Institutet, Department of Clinical Science and Education, Södersjukhuset, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Reno CM, Tanoli T, Bree A, Daphna-Iken D, Cui C, Maloney SE, Wozniak DF, Fisher SJ. Antecedent glycemic control reduces severe hypoglycemia-induced neuronal damage in diabetic rats. Am J Physiol Endocrinol Metab 2013; 304:E1331-7. [PMID: 23592483 PMCID: PMC3680694 DOI: 10.1152/ajpendo.00084.2013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Brain damage due to severe hypoglycemia occurs in insulin-treated people with diabetes. This study tests the hypothesis that chronic insulin therapy that normalizes elevated blood glucose in diabetic rats would be neuroprotective against brain damage induced by an acute episode of severe hypoglycemia. Male Sprague-Dawley rats were split into three groups: 1) control, non-diabetic; 2) STZ-diabetic; and 3) insulin-treated STZ-diabetic. After 3 wk of chronic treatment, unrestrained awake rats underwent acute hyperinsulinemic severe hypoglycemic (10-15 mg/dl) clamps for 1 h. Rats were subsequently analyzed for brain damage and cognitive function. Severe hypoglycemia induced 15-fold more neuronal damage in STZ-diabetic rats compared with nondiabetic rats. Chronic insulin treatment of diabetic rats, which nearly normalized glucose levels, markedly reduced neuronal damage induced by severe hypoglycemia. Fortunately, no cognitive defects associated with the hypoglycemia-induced brain damage were observed in any group. In conclusion, antecedent blood glucose control represents a major modifiable therapeutic intervention that can afford diabetic subjects neuroprotection against severe hypoglycemia-induced brain damage.
Collapse
Affiliation(s)
- Candace M Reno
- Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Kim JH, Jang BG, Choi BY, Kwon LM, Sohn M, Song HK, Suh SW. Zinc chelation reduces hippocampal neurogenesis after pilocarpine-induced seizure. PLoS One 2012; 7:e48543. [PMID: 23119054 PMCID: PMC3485345 DOI: 10.1371/journal.pone.0048543] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 09/26/2012] [Indexed: 12/03/2022] Open
Abstract
Several studies have shown that epileptic seizures increase hippocampal neurogenesis in the adult. However, the mechanism underlying increased neurogenesis after seizures remains largely unknown. Neurogenesis occurs in the subgranular zone (SGZ) of the hippocampus in the adult brain, although an understanding of why it actively occurs in this region has remained elusive. A high level of vesicular zinc is localized in the presynaptic terminals of the SGZ. Previously, we demonstrated that a possible correlation may exist between synaptic zinc localization and high rates of neurogenesis in this area after hypoglycemia. Using a lithium-pilocarpine model, we tested our hypothesis that zinc plays a key role in modulating hippocampal neurogenesis after seizure. Then, we injected the zinc chelator, clioquinol (CQ, 30 mg/kg), into the intraperitoneal space to reduce brain zinc availability. Neuronal death was detected with Fluoro Jade-B and NeuN staining to determine whether CQ has neuroprotective effects after seizure. The total number of degenerating and live neurons was similar in vehicle and in CQ treated rats at 1 week after seizure. Neurogenesis was evaluated using BrdU, Ki67 and doublecortin (DCX) immunostaining 1 week after seizure. The number of BrdU, Ki67 and DCX positive cell was increased after seizure. However, the number of BrdU, Ki67 and DCX positive cells was significantly decreased by CQ treatment. Intracellular zinc chelator, N,N,N0,N-Tetrakis (2-pyridylmethyl) ethylenediamine (TPEN), also reduced seizure-induced neurogenesis in the hippocampus. The present study shows that zinc chelation does not prevent neurodegeneration but does reduce seizure-induced progenitor cell proliferation and neurogenesis. Therefore, this study suggests that zinc has an essential role for modulating hippocampal neurogenesis after seizure.
Collapse
Affiliation(s)
- Jin Hee Kim
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Korea
| | - Bong Geom Jang
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Korea
| | - Bo Young Choi
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Korea
| | - Lyo Min Kwon
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Korea
| | - Min Sohn
- Inha University, Department of Nursing, Incheon, Korea
| | - Hong Ki Song
- Department of Neurology, College of Medicine, Hallym University, Chunchon, Korea
- * E-mail: (HKS); (SWS)
| | - Sang Won Suh
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, Korea
- * E-mail: (HKS); (SWS)
| |
Collapse
|
24
|
Pearson-Leary J, McNay EC. Intrahippocampal administration of amyloid-β(1-42) oligomers acutely impairs spatial working memory, insulin signaling, and hippocampal metabolism. J Alzheimers Dis 2012; 30:413-22. [PMID: 22430529 DOI: 10.3233/jad-2012-112192] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Increasing evidence suggests that abnormal brain accumulation of amyloid-β(1-42) (Aβ(1-42)) oligomers plays a causal role in Alzheimer's disease (AD), and in particular may cause the cognitive deficits that are the hallmark of AD. In vitro, Aβ(1-42) oligomers impair insulin signaling and suppress neural functioning. We previously showed that endogenous insulin signaling is an obligatory component of normal hippocampal function, and that disrupting this signaling led to a rapid impairment of spatial working memory, while delivery of exogenous insulin to the hippocampus enhanced both memory and metabolism; diet-induced insulin resistance both impaired spatial memory and prevented insulin from increasing metabolism or cognitive function. Hence, we tested the hypothesis that Aβ(1-42) oligomers could acutely impair hippocampal metabolic and cognitive processes in vivo in the rat. Our findings support this hypothesis: Aβ(1-42) oligomers impaired spontaneous alternation behavior while preventing the task-associated dip in hippocampal ECF glucose observed in control animals. In addition, Aβ(1-42) oligomers decreased plasma membrane translocation of the insulin-sensitive glucose transporter 4 (GluT4), and impaired insulin signaling as measured by phosphorylation of Akt. These data show in vivo that Aβ(1-42) oligomers can rapidly impair hippocampal cognitive and metabolic processes, and provide support for the hypothesis that elevated Aβ(1-42) leads to cognitive impairment via interference with hippocampal insulin signaling.
Collapse
|
25
|
Darsalia V, Mansouri S, Ortsäter H, Olverling A, Nozadze N, Kappe C, Iverfeldt K, Tracy L, Grankvist N, Sjöholm Å, Patrone C. Glucagon-like peptide-1 receptor activation reduces ischaemic brain damage following stroke in Type 2 diabetic rats. Clin Sci (Lond) 2012; 122:473-83. [PMID: 22150224 PMCID: PMC3268352 DOI: 10.1042/cs20110374] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Revised: 12/05/2011] [Accepted: 12/07/2011] [Indexed: 02/06/2023]
Abstract
Diabetes is a strong risk factor for premature and severe stroke. The GLP-1R (glucagon-like peptide-1 receptor) agonist Ex-4 (exendin-4) is a drug for the treatment of T2D (Type 2 diabetes) that may also have neuroprotective effects. The aim of the present study was to determine the efficacy of Ex-4 against stroke in diabetes by using a diabetic animal model, a drug administration paradigm and a dose that mimics a diabetic patient on Ex-4 therapy. Furthermore, we investigated inflammation and neurogenesis as potential cellular mechanisms underlying the Ex-4 efficacy. A total of seven 9-month-old Type 2 diabetic Goto–Kakizaki rats were treated peripherally for 4 weeks with Ex-4 at 0.1, 1 or 5 μg/kg of body weight before inducing stroke by transient middle cerebral artery occlusion and for 2–4 weeks thereafter. The severity of ischaemic damage was measured by evaluation of stroke volume and by stereological counting of neurons in the striatum and cortex. We also quantitatively evaluated stroke-induced inflammation, stem cell proliferation and neurogenesis. We show a profound anti-stroke efficacy of the clinical dose of Ex-4 in diabetic rats, an arrested microglia infiltration and an increase of stroke-induced neural stem cell proliferation and neuroblast formation, while stroke-induced neurogenesis was not affected by Ex-4. The results show a pronounced anti-stroke, neuroprotective and anti-inflammatory effect of peripheral and chronic Ex-4 treatment in middle-aged diabetic animals in a preclinical setting that has the potential to mimic the clinical treatment. Our results should provide strong impetus to further investigate GLP-1R agonists for their neuroprotective action in diabetes, and for their possible use as anti-stroke medication in non-diabetic conditions.
Collapse
Key Words
- exendin-4 (ex-4)
- goto–kakizaki (gk) rat
- middle cerebral artery occlusion (mcao)
- neurogenesis
- neuroprotection
- brdu, bromodeoxyuridine
- bw, body weight
- cns, central nervous system
- dapi, 4′,6-diamidino-2-phenylindole
- dcx, doublecortin
- ex-4, exendin-4
- gk, goto–kakizaki
- glp-1r, glucagon-like peptide-1 receptor
- ihc, immunohistochemistry
- mca, middle cerebral artery
- mcao, mca occlusion
- svz, subventricular zone
- t2d, type 2 diabetes
Collapse
Affiliation(s)
- Vladimer Darsalia
- *Diabetes Research Unit, Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Shiva Mansouri
- *Diabetes Research Unit, Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Ortsäter
- *Diabetes Research Unit, Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Anna Olverling
- *Diabetes Research Unit, Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Nino Nozadze
- *Diabetes Research Unit, Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Camilla Kappe
- *Diabetes Research Unit, Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Kerstin Iverfeldt
- †Department of Neurochemistry, Stockholm University, Stockholm, Sweden
| | - Linda M. Tracy
- †Department of Neurochemistry, Stockholm University, Stockholm, Sweden
| | - Nina Grankvist
- *Diabetes Research Unit, Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Åke Sjöholm
- *Diabetes Research Unit, Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Cesare Patrone
- *Diabetes Research Unit, Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
26
|
Mansouri S, Ortsäter H, Pintor Gallego O, Darsalia V, Sjöholm A, Patrone C. Pituitary adenylate cyclase-activating polypeptide counteracts the impaired adult neural stem cell viability induced by palmitate. J Neurosci Res 2011; 90:759-68. [PMID: 22183970 DOI: 10.1002/jnr.22803] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 08/30/2011] [Indexed: 12/30/2022]
Abstract
Diabetes and obesity are characterized by hyperlipidemia and represent risk factors for premature neurological disorders. Diabetic/obese animals have impaired adult neurogenesis. We hypothesize that lipotoxicity leading to neurogenesis impairment plays a role in the development of neurological complications. If so, normalizing neurogenesis in diabetes/obesity could be therapeutically useful in counteracting neurological dysfunction. The goal of this study was to determine the potential of pituitary adenylate cyclase-activating polypeptide (PACAP) to protect adult neural stem cells (NSCs) from lipotoxicity and to study the expression of PACAP receptors in NSCs under lipotoxic conditions in vitro and in the subventricular zone in vivo. The viability of NSCs isolated from the adult mouse brain subventricular zone was assessed in the presence of a high-fat milieu, as mimicked by palmitate, which characterizes diabetic lipotoxicity. Regulation studies of PACAP receptors were performed by quantitative PCR on NSCs in vitro or on subventricular tissues isolated from obese ob/ob mice and their lean littermates. We show that palmitate impairs NSC viability by promoting lipoapoptosis. We also show that PACAP counteracts lipotoxicity via PAC-1 receptor activation. Studies on PACAP receptor expression revealed that PAC-1 and VPAC-2 are expressed by NSC in vitro and are upregulated by palmitate treatment and that PAC-1, VPAC-1, and VPAC-2 are expressed in the subventricular zone/striatum in vivo and are upregulated in ob/ob mice. The present study reveals a previously uncharacterized role of PACAP to protect NSC from lipotoxicity and suggests a potential therapeutic role for PACAP receptor agonists in the treatment of neurological complications in obesity and diabetes.
Collapse
Affiliation(s)
- Shiva Mansouri
- Diabetes Research Unit, Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
27
|
Takeda A, Sakamoto K, Tamano H, Fukura K, Inui N, Suh SW, Won SJ, Yokogoshi H. Facilitated neurogenesis in the developing hippocampus after intake of theanine, an amino acid in tea leaves, and object recognition memory. Cell Mol Neurobiol 2011; 31:1079-88. [PMID: 21604187 DOI: 10.1007/s10571-011-9707-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 05/05/2011] [Indexed: 12/28/2022]
Abstract
Theanine, γ-glutamylethylamide, is one of the major amino acid components in green tea. In this study, cognitive function and the related mechanism were examined in theanine-administered young rats. Newborn rats were fed theanine through dams, which were fed water containing 0.3% theanine, and then fed water containing 0.3% theanine after weaning. Theanine level in the brain was under the detectable limit 6 weeks after the start of theanine administration. Theanine administration did not influence locomotor activity in the open-field test. However, rearing behavior was significantly increased in theanine-administered rats, suggesting that exploratory activity is increased by theanine intake. Furthermore, object recognition memory was enhanced in theanine-administered rats. The increase in exploratory activity in the open-field test seems to be associated with the enhanced object recognition memory after theanine administration. On the other hand, long-term potentiation (LTP) induction at the perforant path-granule cell synapse was not changed by theanine administration. To check hippocampal neurogenesis, BrdU was injected into rats 3 weeks after the start of theanine administration, and brain-derived neurotropic factor (BDNF) level was significantly increased at this time. Theanine intake significantly increased the number of BrdU-, Ki67-, and DCX-labeled cells in the granule cell layer 6 weeks after the start of theanine administration. This study indicates that 0.3% theanine administration facilitates neurogenesis in the developing hippocampus followed by enhanced recognition memory. Theanine intake may be of benefit to the postnatal development of hippocampal function.
Collapse
Affiliation(s)
- Atsushi Takeda
- Graduate School of Pharmaceutical Sciences, University of Shizuoka, Global COE, Shizuoka, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Silverstein JM, Musikantow D, Puente EC, Daphna-Iken D, Bree AJ, Fisher SJ. Pharmacologic amelioration of severe hypoglycemia-induced neuronal damage. Neurosci Lett 2011; 492:23-8. [PMID: 21272612 DOI: 10.1016/j.neulet.2011.01.045] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 01/19/2011] [Indexed: 10/18/2022]
Abstract
Hypoglycemia is a common complication for insulin treated people with diabetes. Severe hypoglycemia, which occurs in the setting of excess or ill-timed insulin administration, has been shown to cause brain damage. Previous pre-clinical studies have shown that memantine (an N-methyl-d-aspartate receptor antagonist) and erythropoietin can be neuroprotective in other models of brain injury. We hypothesized that these agents might also be neuroprotective in response to severe hypoglycemia-induced brain damage. To test this hypothesis, 9-week old, awake, male Sprague-Dawley rats underwent hyperinsulinemic (0.2 U kg(-1)min(-1)) hypoglycemic clamps to induce severe hypoglycemia (blood glucose 10-15 mg/dl for 90 min). Animals were randomized into control (vehicle) or pharmacological treatments (memantine or erythropoietin). One week after severe hypoglycemia, neuronal damage was assessed by Fluoro-Jade B and hematoxylin and eosin staining of brain sections. Treatment with both memantine and erythropoietin significantly decreased severe hypoglycemia-induced neuronal damage in the cortex by 35% and 39%, respectively (both p<0.05 vs. controls). These findings demonstrate that memantine and erythropoietin provide a protective effect against severe hypoglycemia-induced neuronal damage.
Collapse
Affiliation(s)
- Julie M Silverstein
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine,Washington University, St. Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
29
|
Rafalski VA, Brunet A. Energy metabolism in adult neural stem cell fate. Prog Neurobiol 2010; 93:182-203. [PMID: 21056618 DOI: 10.1016/j.pneurobio.2010.10.007] [Citation(s) in RCA: 218] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 10/20/2010] [Accepted: 10/28/2010] [Indexed: 12/26/2022]
Abstract
The adult mammalian brain contains a population of neural stem cells that can give rise to neurons, astrocytes, and oligodendrocytes and are thought to be involved in certain forms of memory, behavior, and brain injury repair. Neural stem cell properties, such as self-renewal and multipotency, are modulated by both cell-intrinsic and cell-extrinsic factors. Emerging evidence suggests that energy metabolism is an important regulator of neural stem cell function. Molecules and signaling pathways that sense and influence energy metabolism, including insulin/insulin-like growth factor I (IGF-1)-FoxO and insulin/IGF-1-mTOR signaling, AMP-activated protein kinase (AMPK), SIRT1, and hypoxia-inducible factors, are now implicated in neural stem cell biology. Furthermore, these signaling modules are likely to cooperate with other pathways involved in stem cell maintenance and differentiation. This review summarizes the current understanding of how cellular and systemic energy metabolism regulate neural stem cell fate. The known consequences of dietary restriction, exercise, aging, and pathologies with deregulated energy metabolism for neural stem cells and their differentiated progeny will also be discussed. A better understanding of how neural stem cells are influenced by changes in energy availability will help unravel the complex nature of neural stem cell biology in both the normal and diseased state.
Collapse
|
30
|
Abstract
In the adult brain, neurogenesis occurs in the subgranular zone of the dentate gyrus (DG), where high levels of vesicular zinc are localized in the presynaptic terminals. To determine whether zinc has a role in modulating hippocampal neurogenesis under normal or pathologic conditions, we manipulated the level of vesicular zinc experimentally. To reduce hippocampal vesicular zinc, rats were either fed a zinc-deficient diet or treated with a zinc chelator, clioquinol (CQ). The number of progenitor cells and immature neurons was decreased significantly in the DG after 6 weeks of dietary zinc deprivation. Conversely, the number of progenitor cells and immature neurons was restored after a 2-week reversal to a normal zinc-containing diet. Similarly, a 1-week treatment with the zinc chelator, CQ, reduced the number of progenitor cells. The results of our previous study showed that hypoglycemia increased hippocampal neurogenesis. This study shows that zinc chelation reduced hypoglycemia-induced progenitor cell proliferation and neurogenesis. Finally, the role of vesicular zinc on neurogenesis was further assessed in zinc transporter 3 (ZnT3) gene deleted mice. Zinc transporter 3 knockout (KO) mice had significantly fewer proliferating progenitor cells and immature neurons after hypoglycemia. Our data provide converging evidence in support of the essential role zinc has in modulating hippocampal neurogenesis.
Collapse
|
31
|
Wang SH, Sun ZL, Guo YJ, Yuan Y, Yang BQ. Diabetes impairs hippocampal function via advanced glycation end product mediated new neuron generation in animals with diabetes-related depression. Toxicol Sci 2009; 111:72-9. [PMID: 19502549 DOI: 10.1093/toxsci/kfp126] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The diabetes-induced reduction of neurogenesis in hippocampal dentate and its reversal with antidepressant medications implies a potential mechanism for diabetes-related depression and cognitive decline. In the following article, the role of advanced glycation end products (AGEs) in hippocampal neurogenesis deficits in diabetic animals with depression has been further explained in the light of an in vitro study. Diabetes was induced in animals with the use of streptozotocin (55 mg/kg, i.p.), and the animals then divided into those with and those without depression-like behaviors as analyzed by behavioral tests. The AGE formation inhibitor aminoguanidine (10 mg/kg) was administrated for an additional 4 weeks. Proliferating cells, their survival, and their phenotype fate were monitored with bromodeoxyuridine labeling and confocal laser microscopy. The presence of AGE peptides was determined with the use of a flow injection assay. Animals with diabetes and depressive symptoms displayed a reduction in hippocampal neurogenesis and an elevated serum level of AGE peptides, both of which were reversed by a 4-week regimen of aminoguanidine (10 mg/kg, i.p.), which inhibits AGE formation; in addition, the depressive behaviors were improved. These findings provided in vivo evidence that diabetes impairs hippocampal function via the AGE-mediated generation of new neurons. This likely represents a putative mechanism that is responsible for diabetes-related depression and cognitive decline, and it suggests a potential approach for future research.
Collapse
Affiliation(s)
- Shao-hua Wang
- The Department of Endocrinology, affiliated ZhongDa Hospital of Southeast University, Nanjing, PR China 210009
| | | | | | | | | |
Collapse
|
32
|
Estrada FS, Hernandez VS, Medina MP, Corona-Morales AA, Gonzalez-Perez O, Vega-Gonzalez A, Zhang L. Astrogliosis is temporally correlated with enhanced neurogenesis in adult rat hippocampus following a glucoprivic insult. Neurosci Lett 2009; 459:109-14. [PMID: 19446003 DOI: 10.1016/j.neulet.2009.05.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Revised: 04/23/2009] [Accepted: 05/06/2009] [Indexed: 11/28/2022]
Abstract
2-Deoxy-d-glucose (2-DG) administration causes transient depletion of glucose derivates and ATP. Hence, it can be used in a model system to study the effects of a mild glycoprivic brain insult mimicking transient hypoglycemia, which often occurs when insulin or oral hypoglycemic agents are administered for diabetes control. In the present study, the effect of a single 2-DG application (500mg/kg, a clinically applicable dose) on glial reactivity and neurogenesis in adult rat hippocampus was examined, as well as a possible temporal correlation between these two phenomena. Post-insult (PI) glial reactivity time course was assessed by immunoreaction against glial-fibrillary acidic protein (GFAP) during the following 5 consecutive days. A clear increase of GFAP immunoreactivity in hilus was observed from 48 to 96h PI. Moreover, enhanced labeling of long radial processes in the granule cell layer adjacent to hilus was evidenced. On the other hand, a transient increase of progenitor cell proliferation was detected in the subgranular zone, prominently at 48h PI, coinciding with the temporal peak of glial activation. This increase resulted in an augment of neuroblasts double labeled with 5-bromo-deoxyuridine (BrdU) and with double cortin (DCX) at day 7 PI. Around half of these cells survived 28 days showing matured neuronal phenotype double labeled by BrdU and a neuronal specific nuclear protein marker (NeuN). These findings suggest that a transient neuroglycoprivic state exerts a short-term effect on glial activation that possibly triggers a long-term effect on neurogenesis in hippocampus.
Collapse
Affiliation(s)
- Felipe S Estrada
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, México D. F., 04510, Mexico
| | | | | | | | | | | | | |
Collapse
|
33
|
Kosuge Y, Imai T, Kawaguchi M, Kihara T, Ishige K, Ito Y. Subregion-specific vulnerability to endoplasmic reticulum stress-induced neurotoxicity in rat hippocampal neurons. Neurochem Int 2008; 52:1204-11. [DOI: 10.1016/j.neuint.2007.12.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Revised: 12/14/2007] [Accepted: 12/19/2007] [Indexed: 12/13/2022]
|
34
|
Beauquis J, Saravia F, Coulaud J, Roig P, Dardenne M, Homo-Delarche F, De Nicola A. Prominently decreased hippocampal neurogenesis in a spontaneous model of type 1 diabetes, the nonobese diabetic mouse. Exp Neurol 2008; 210:359-67. [DOI: 10.1016/j.expneurol.2007.11.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2007] [Revised: 11/08/2007] [Accepted: 11/09/2007] [Indexed: 11/24/2022]
|
35
|
Jauch-Chara K, Hallschmid M, Gais S, Schmid SM, Oltmanns KM, Colmorgen C, Born J, Schultes B. Hypoglycemia during sleep impairs consolidation of declarative memory in type 1 diabetic and healthy humans. Diabetes Care 2007; 30:2040-5. [PMID: 17468346 DOI: 10.2337/dc07-0067] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Early nocturnal sleep enhances the consolidation of declarative memories acquired during prior wakefulness. Patients with type 1 diabetes frequently experience hypoglycemic episodes during sleep. We investigated whether short-lasting hypoglycemia during early nocturnal sleep affects the sleep-associated consolidation of declarative memories. RESEARCH DESIGN AND METHODS Sixteen type 1 diabetic patients and 16 healthy subjects matched for age and BMI were tested. On one condition, a linear fall of plasma glucose to 2.2 mmol/l was induced within 60 min by infusing insulin during early sleep. On the control condition, euglycemia (>3.86 mmol/l) was maintained throughout the night. In the morning, subjects recalled word pairs learned in the preceding evening. To assess mood and attention, a symptom questionnaire, an adjective check list, and the Stroop test were applied. Also, auditory event-related brain potentials were recorded. RESULTS After euglycemia, subjects recalled 1.5 +/- 0.5 more word pairs than after hypoglycemia (P < 0.01), remembering 2.0 +/- 0.6 more word pairs than at immediate recall before sleep (P = 0.002). Across the hypoglycemic night, no such gain occurred (+0.5 +/- 0.6 words; P = 0.41). Hypoglycemia during sleep also impaired mood (P < 0.05) but did not affect attention. Effects compared well between type 1 diabetic patients and healthy control subjects. CONCLUSIONS Our findings indicate specific sensitivity of declarative memory consolidation during sleep to rather short episodes of mild hypoglycemia. This effect may disable memory processing in type 1 diabetic patients prone to nocturnal hypoglycemic episodes and underlines the importance of considering sleep as a critical period in the treatment of these patients.
Collapse
|
36
|
Hong SM, Liu Z, Fan Y, Neumann M, Won SJ, Lac D, Lum X, Weinstein PR, Liu J. Reduced hippocampal neurogenesis and skill reaching performance in adult Emx1 mutant mice. Exp Neurol 2007; 206:24-32. [PMID: 17490651 DOI: 10.1016/j.expneurol.2007.03.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2007] [Revised: 02/14/2007] [Accepted: 03/13/2007] [Indexed: 12/16/2022]
Abstract
Mammalian homeobox gene Emx family is involved in the development of the rostral brain. Loss-of-function studies suggest that, despite the agenesis of corpus callosum, the Emx1 mutants display relatively modest defects compared to the Emx2 mutants. However, the role of the Emx1 in neurogenesis and brain function has never been explored. We used unbiased stereology to determine the number of proliferating progenitors and immature neurons in the adult neurogenic zones. Although previous studies have established that the formation of the dentate gyrus (DG) requires Emx2, we found that the adult Emx1 mutants also exhibited a smaller DG, reduced number of proliferating progenitor cells and immature neurons in the DG, in contrast to the indistinguishable level of neurogenesis in the subventricular zone when compared to the wild type mice. In view of the involvement of callosal projection neurons in mediating interhemispheric crosstalk and spatial coupling between the limbs, and the importance of DG in hippocampus-dependent function in learning and memory, we assessed motor and cognitive functions. Emx1 deletion impaired performance on a forelimb skill reaching task and attenuated training induced hippocampal neurogenesis, but it did not affect motor activity or basic motor function as evaluated in the open field, wire hanging and rotor rod tests. Unexpectedly, the adult Emx1 mutant mice did not exhibit impairment in spatial learning and memory in the Barnes maze test. Our data suggest that deletion of the Emx1 gene reduces hippocampal neurogenesis and affects higher motor function that requires extensive learning.
Collapse
Affiliation(s)
- Shwuhuey M Hong
- Department of Neurological Surgery (112C), University of California at San Francisco, UCSF and SFVAMC, 4150 Clement Street, San Francisco, CA 94121, USA, and Department of Neurosurgery, HuaShan Hospital, Shanghai, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Fan Y, Liu Z, Weinstein PR, Fike JR, Liu J. Environmental enrichment enhances neurogenesis and improves functional outcome after cranial irradiation. Eur J Neurosci 2007; 25:38-46. [PMID: 17241265 DOI: 10.1111/j.1460-9568.2006.05269.x] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Radiation therapy is a widely used treatment for brain tumors but it can cause delayed progressive cognitive decline and memory deficits. Previous studies suggested that this neurocognitive dysfunction might be linked to the impairment of hippocampal neurogenesis. However, little is known regarding how to reduce the cognitive impairment caused by radiation therapy. To investigate whether environmental enrichment (EE) promotes neurogenesis and cognitive function after irradiation, irradiated gerbils were housed in EE for 2 months and evaluated by neurobehavioral testing for learning and memory function, and immunohistochemical analysis for neurogenesis. Our results demonstrated that even relatively low doses (5-10 Gy) of irradiation could acutely abolish precursor cell proliferation in the dentate gyrus by more than 90%. This reduction in precursor proliferation was persistent and led to a significant decline in the granule cell population 9 months later. EE housing enhanced the number of newborn neurons and increased residual neurogenesis. EE also significantly increased the total number of immature neurons in the dentate gyrus. Furthermore, irradiated animals after EE housing showed a significant improvement in spatial learning and memory during the water-maze test and in rotorod motor learning over a 5-day training paradigm. In conclusion, EE has a positive impact on hippocampal neurogenesis and functional recovery in irradiated adult gerbils. Our data suggest that there is still a considerable amount of plasticity remaining in the hippocampal progenitor cells in adult animals after radiation injury, which can become a target of therapeutic intervention for radiation-induced cognitive dysfunction.
Collapse
Affiliation(s)
- Yang Fan
- Department of Neurological Surgery (112C), University of California at San Francisco, San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | | | | | | | | |
Collapse
|
38
|
Hayes RP, Nakano M, Muchmore D, Schmitke J. Effect of standard (self-directed) training versus intensive training for Lilly/Alkermes human insulin inhalation powder delivery system on patient-reported outcomes and patient evaluation of the system. Diabetes Technol Ther 2007; 9:89-98. [PMID: 17316103 DOI: 10.1089/dia.2006.0043] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND Inhaled insulin may provide patients with diabetes a safe, efficacious method of insulin delivery without the burden of injection, but complexity of and time required for training in proper use of delivery systems have not been evaluated. METHODS This 4-week, multicenter, single-blind, randomized parallel-group study compared the effect of self-directed [written text-graphic directions for use (DFU) with patient-assistance phone number] or intensive (same DFU, personal training by study personnel, inspiratory flow rate coaching) training for the Lilly/Alkermes human insulin inhalation powder (HIIP) delivery system on patient-reported outcomes (PROs). Patients with type 2 diabetes poorly controlled on oral therapy (n = 102, mean hemoglobin A1C = 9.3%) were administered measures of vitality, diabetes-associated symptoms, fear of hypoglycemia, insulin-delivery system satisfaction, and a delivery system-specific evaluation questionnaire. Analysis of covariance models were used to compare the effect on PROs of treatment of diabetes for 1 month following the two training methods. Paired t tests were used to determine change in PROs after treatment with HIIP. RESULTS PROs did not differ significantly between training groups. Patients in both groups positively evaluated the delivery system, but the intensive group agreed significantly (P < 0.05) more strongly that the DFU was easy to follow. Improvements in vitality and symptoms of fatigue and increases in fear of hypoglycemia were detected among all patients after using HIIP (P < 0.05). CONCLUSION Training for this HIIP delivery system can be self-directed without detrimental effects on PROs, making it potentially a more patient-friendly insulin-delivery method that should appeal to both clinicians and patients.
Collapse
Affiliation(s)
- Risa P Hayes
- Global Health Outcomes, Eli Lilly and Company, Indianapolis, Indiana 46285, USA.
| | | | | | | |
Collapse
|
39
|
Parent JM. Adult neurogenesis in the intact and epileptic dentate gyrus. PROGRESS IN BRAIN RESEARCH 2007; 163:529-40. [PMID: 17765736 DOI: 10.1016/s0079-6123(07)63028-3] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neurogenesis persists throughout life in the adult mammalian dentate gyrus. Adult-born dentate granule cells integrate into existing hippocampal circuitry and may provide network plasticity necessary for certain forms of hippocampus-dependent learning and memory. Neural stem cells and neurogenesis in the adult dentate gyrus are regulated by a variety of environmental, physiological, and molecular factors. These include aging, stress, exercise, neurovascular components of the stem cell niche, growth factors, neurotransmitters, and hormones. Seizure activity also influences dentate granule cell neurogenesis. Production of adult-born neurons increases in rodent models of temporal lobe epilepsy, and both newborn and pre-existing granule neurons contribute to aberrant axonal reorganization in the epileptic hippocampus. Prolonged seizures also disrupt the migration of dentate granule cell progenitors and lead to hilar-ectopic granule cells. The ectopic granule neurons appear to integrate abnormally and contribute to network hyperexcitability. Similar findings of granule cell layer dispersion and ectopic granule neurons in human TLE suggest that aberrant neurogenesis contributes to epileptogenesis or learning and memory disturbances in this epilepsy syndrome.
Collapse
Affiliation(s)
- Jack M Parent
- Department of Neurology, University of Michigan Medical Center, Ann Arbor, MI 48109-2200, USA.
| |
Collapse
|
40
|
Ballok DA. Neuroimmunopathology in a murine model of neuropsychiatric lupus. ACTA ACUST UNITED AC 2006; 54:67-79. [PMID: 17223198 PMCID: PMC2577581 DOI: 10.1016/j.brainresrev.2006.12.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2006] [Revised: 12/09/2006] [Accepted: 12/11/2006] [Indexed: 01/18/2023]
Abstract
Animal models are extremely useful tools in defining pathogenesis and treatment of human disease. For many years researchers believed that structural damage to the brain of neuropsychiatric (NP) patients lead to abnormal mental function, but this possibility was not extensively explored until recently. Imaging studies of NP-systemic lupus erythematosus (SLE) support the notion that brain cell death accounts for the emergence of neurologic and psychiatric symptoms, and evidence suggests that it is an autoimmunity-induced brain disorder characterized by profound metabolic alterations and progressive neuronal loss. While there are a number of murine models of SLE, this article reviews recent literature on the immunological connections to neurodegeneration and behavioral dysfunction in the Fas-deficient MRL model of NP-SLE. Probable links between spontaneous peripheral immune activation, the subsequent central autoimmune/inflammatory responses in MRL/MpJ-Tnfrsf6(lpr) (MRL-lpr) mice and the sequential mode of events leading to Fas-independent neurodegenerative autoimmune-induced encephalitis will be reviewed. The role of hormones, alternative mechanisms of cell death, the impact of central dopaminergic degeneration on behavior, and germinal layer lesions on developmental/regenerative capacity of MRL-lpr brains will also be explored. This model can provide direction for future therapeutic interventions in patients with this complex neuroimmunological syndrome.
Collapse
Affiliation(s)
- David A Ballok
- Department of Psychiatry and Behavioral Neurosciences, HSC Rm 4N4, McMaster University, 1200 Main St. West, Hamilton, Ontario, Canada L8N 3Z5.
| |
Collapse
|
41
|
Hayes RP, Bowman L, Monahan PO, Marrero DG, McHorney CA. Understanding diabetes medications from the perspective of patients with type 2 diabetes: prerequisite to medication concordance. DIABETES EDUCATOR 2006; 32:404-14. [PMID: 16772656 DOI: 10.1177/0145721706288182] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE The purpose of this qualitative study was to explore diabetes medication experiences of patients with type 2 diabetes. METHODS Study participants were recruited through newspaper advertisements and letters from primary and specialty clinics in Indianapolis, Indiana. The same professional facilitator conducted 18 focus groups (7 male, 11 female) at a Veteran Affairs facility in Indianapolis, Indiana. Participants were 138 socioeconomically diverse individuals with type 2 diabetes (68% female, 74% > or = 50 years old, 61% non-Hispanic Caucasian). Participants were on a variety of diabetes medication regimens (61% injectable insulin monotherapy or in combination with orals). Content analysis of focus group transcripts was used to establish themes. RESULTS Three major themes emerged across the focus groups: (1) the inconvenience and inflexibility of the timing and frequency of administration of many diabetes treatments on participants' lives, (2) the desire to avoid injections and/or insulin therapy, and (3) the physical and emotional side effects of diabetes medications. Assuming equivalent effectiveness of 2 medications, participants would weigh the convenience and flexibility, route of administration, side effects, and cost of the medications to arrive at a treatment preference. Participants believed they currently had no opportunity to express their treatment preference to their health care provider. CONCLUSIONS The best therapeutic option for a patient with type 2 diabetes will be one that is clinically effective and consistent with the patient's needs and preferences. The challenge for diabetes educators is to support patients in articulating and incorporating their needs and preferences into the treatment decision-making process.
Collapse
Affiliation(s)
- Risa P Hayes
- Global Health Outcomes Research, Eli Lilly & Company, Indianapolis, Indiana (Dr Hayes, Dr Bowman)
| | - Lee Bowman
- Global Health Outcomes Research, Eli Lilly & Company, Indianapolis, Indiana (Dr Hayes, Dr Bowman)
| | - Patrick O Monahan
- Division of Biostatistics, Department of Medicine, Indiana University School of Medicine, Indianapolis (Dr Monahan)
| | - David G Marrero
- Division of Endocrinology & Metabolism, Indiana University School of Medicine, Indianapolis (Dr Marrero)
| | - Colleen A McHorney
- Outcomes Research & Management, Merck & Company, West Point, Pennsylvania (Dr McHorney)
| |
Collapse
|
42
|
Abstract
The recent identification of endogenous neural stem cells and persistent neuronal production in the adult brain suggests a previously unrecognized capacity for self-repair after brain injury. Neurogenesis not only continues in discrete regions of the adult mammalian brain, but new evidence also suggests that neural progenitors form new neurons that integrate into existing circuitry after certain forms of brain injury in the adult. Experimental stroke in adult rodents and primates increases neurogenesis in the persistent forebrain subventricular and hippocampal dentate gyrus germinative zones. Of greater relevance for regenerative potential, ischemic insults stimulate endogenous neural progenitors to migrate to areas of damage and form neurons in otherwise dormant forebrain regions, such as the neostriatum and hippocampal pyramidal cell layer, of the mature brain. This review summarizes the current understanding of adult neurogenesis and its regulation in vivo, and describes evidence for stroke-induced neurogenesis and neuronal replacement in the adult. Current strategies used to modify endogenous neurogenesis after ischemic brain injury also will be discussed, as well as future research directions with potential for achieving regeneration after stroke and other brain insults.
Collapse
Affiliation(s)
- Robin J Lichtenwalner
- Department of Neurology, University of Michigan Medical Center, Ann Arbor, Michigan 48109-0585, USA
| | | |
Collapse
|
43
|
Matsumori Y, Hong SM, Fan Y, Kayama T, Hsu CY, Weinstein PR, Liu J. Enriched environment and spatial learning enhance hippocampal neurogenesis and salvages ischemic penumbra after focal cerebral ischemia. Neurobiol Dis 2005; 22:187-98. [PMID: 16361108 DOI: 10.1016/j.nbd.2005.10.015] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2005] [Revised: 10/25/2005] [Accepted: 10/29/2005] [Indexed: 01/17/2023] Open
Abstract
Enriched environment (EE) has been shown to increase neurogenesis in the adult brain. The aim of this study is to determine the effect of EE and spatial learning on neurogenesis following ischemic stroke. Male adult SD rats were subjected to sham surgery or distal middle cerebral artery occlusion (MCAO). MCAO induced a transient increase followed by a sustained depression of progenitor cell proliferation and neuroblast production below baseline level in both ipsilateral and contralateral DG compared to sham. Increased neuronal differentiation and neurogenesis in the DG were observed in both sham and MCAO rats following 8 weeks in the EE combined with spatial learning, compared to rats housed in the standard environment. EE/Learning also restored the total number of neuroblasts in the DG after MCAO compared to sham. Furthermore, EE/learning enhanced the density of NeuN positive cells in the ischemic penumbra, though no new neurons were detected in this region.
Collapse
Affiliation(s)
- Yasuhiko Matsumori
- Department of Neurological Surgery (112C), University of California, San Francisco, CA 94121, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Current literature in diabetes. Diabetes Metab Res Rev 2005; 21:560-7. [PMID: 16240284 DOI: 10.1002/dmrr.604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
45
|
Rankins D, Wellard RM, Cameron F, McDonnell C, Northam E. The impact of acute hypoglycemia on neuropsychological and neurometabolite profiles in children with type 1 diabetes. Diabetes Care 2005; 28:2771-3. [PMID: 16249555 DOI: 10.2337/diacare.28.11.2771] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Debbie Rankins
- Australian Centre for Child Neuropsychological Studies, Murdoch Childrens Research Institute, Parkville, Victoria, Australia
| | | | | | | | | |
Collapse
|
46
|
Abstract
Hot flashes have a negative influence on the quality of life in hypogonadal women. There seem to be many regulators of hot flashes that add to the complexity of these profound vasomotor symptoms. In this paper, focus is placed on selected factors that may be involved in the relationship between hot flashes and cognitive status. During the hypogonadal state, the brain becomes relatively deprived of glucose. Hot flashes were found to be associated with transient inadequacies in brain levels of glucose. Neuroglucopenia was shown to initiate a cascade of immediate and delayed reactions leading to neuronal damage. Estrogen is known to diminish neuroglucopenia. In a pilot study on postmenopausal women, it was observed that cognitive performance of women who reported having hot flashes during their menopausal years was better than in women who did not experience hot flashes. It is hypothesized that a hot flash triggers a counterregulatory mechanism of the central nervous system to the insufficiency of glucose delivery to the brain and may have a beneficial effect on cognitive health in postmenopausal women. Possible therapeutic implications of this hypothesis are discussed in the light of current knowledge.
Collapse
Affiliation(s)
- Anna Ratka
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA.
| |
Collapse
|