1
|
Wang F, Han X, Mu Q, Chen H, Wu Y, Kang Y, Liu Y. Cerebrospinal fluid mesencephalic astrocyte-derived neurotrophic factor: A moderating effect on sleep time and cognitive function. J Psychiatr Res 2024; 176:33-39. [PMID: 38838432 DOI: 10.1016/j.jpsychires.2024.05.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/15/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Sleeping late has been associated with cognitive impairment, and insufficient sleep can affect the secretion of feeding-related cytokines. Feeding-related cytokines may contribute to cognitive deficits resulting from delayed bedtime. Glial cell line-derived neurotrophic factor (GDNF) and mesencephalic astrocyte-derived neurotrophic factor (MANF), which are feeding-related neurotrophic factors, have been associated with improved cognitive function and neuroprotective abilities. Enhanced expression of GDNF and MANF is linked to increased energy expenditure and hyperphagia, respectively. AIMS This study aimed to investigate the association between cerebrospinal fluid (CSF) GDNF, MANF, cognition, and sleep time and to explore the moderating effects of GDNF and MANF on cognitive impairment in individuals who sleep late. METHOD This cross-sectional study included participants (mean age 31.76 ± 10.22 years) who were categorized as ≤23 o'clock sleepers (n = 66) and >23 o'clock sleepers (n = 125) based on sleep time. Cognition was assessed using Montreal Cognitive Assessment (MoCA), and GDNF and MANF levels in CSF were measured. RESULTS MANF may play a moderating role in the relationship between sleep time and cognition (R2 = 0.06, β = 0.59, p = 0.031). Age showed a negative correlation with MoCA scores (R2 = 0.08, β = -0.18), while education exhibited a positive correlation (β = 0.17, both p < 0.05). Only ≤23 o'clock sleepers exhibited a negative correlation between MANF levels and BMI (r = -0.35, p = 0.005). CONCLUSIONS This study provides hitherto undocumented evidence of the potential protective effect of CSF MANF on cognitive impairment of late sleepers, which suggests that maintaining a regular sleep schedule may contribute to cognition and overall health, with MANF playing a role in this process.
Collapse
Affiliation(s)
- Fan Wang
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, 100096, China; Xinjiang Key Laboratory of Neurological Disorder Research, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830063, China.
| | - Xiaoli Han
- Clinical Nutrition Department, Friendship Hospital, Urumqi, 830049, China
| | - Qingshuang Mu
- Xinjiang Key Laboratory of Neurological Disorder Research, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830063, China
| | - Hongxu Chen
- Xinjiang Key Laboratory of Neurological Disorder Research, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830063, China
| | - Yan Wu
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, 100096, China
| | - Yimin Kang
- Medical Neurobiology Lab, Inner Mongolia Medical University, Huhhot, 010110, China
| | - Yanlong Liu
- School of Mental Health, Wenzhou Medical University, Wenzhou, 325035, China; Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
2
|
Montaño-Rodriguez AR, Schorling T, Andressoo JO. Striatal GDNF Neurons Chemoattract RET-Positive Dopamine Axons at Seven Times Farther Distance Than Medium Spiny Neurons. Cells 2024; 13:1059. [PMID: 38920687 PMCID: PMC11202212 DOI: 10.3390/cells13121059] [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: 05/16/2024] [Revised: 06/08/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is among the strongest dopamine neuron function- and survival-promoting factors known. Due to this reason, it has clinical relevance in dopamine disorders such as Parkinson's disease and schizophrenia. In the striatum, GDNF is exclusively expressed in interneurons, which make up only about 0.6% of striatal cells. Despite clinical significance, histological analysis of striatal GDNF system arborization and relevance to incoming dopamine axons, which bear its receptor RET, has remained enigmatic. This is mainly due to the lack of antibodies able to visualize GDNF- and RET-positive cellular processes; here, we overcome this problem by using knock-in marker alleles. We find that GDNF neurons chemoattract RET+ axons at least seven times farther in distance than medium spiny neurons (MSNs), which make up 95% of striatal neurons. Furthermore, we provide evidence that tyrosine hydroxylase, the rate-limiting enzyme in dopamine synthesis, is enriched towards GDNF neurons in the dopamine axons. Finally, we find that GDNF neuron arborizations occupy approximately only twelve times less striatal volume than 135 times more abundant MSNs. Collectively, our results improve our understanding of how endogenous GDNF affects striatal dopamine system function.
Collapse
Affiliation(s)
- Ana Rosa Montaño-Rodriguez
- Department of Pharmacology, Faculty of Medicine, Helsinki Institute of Life Science, University of Helsinki, 00290 Helsinki, Finland; (A.R.M.-R.); (T.S.)
| | - Tabea Schorling
- Department of Pharmacology, Faculty of Medicine, Helsinki Institute of Life Science, University of Helsinki, 00290 Helsinki, Finland; (A.R.M.-R.); (T.S.)
| | - Jaan-Olle Andressoo
- Department of Pharmacology, Faculty of Medicine, Helsinki Institute of Life Science, University of Helsinki, 00290 Helsinki, Finland; (A.R.M.-R.); (T.S.)
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, 17177 Stockholm, Sweden
| |
Collapse
|
3
|
Aldhshan MS, Mizuno TM. Environmental enrichment accentuates glucose-induced feeding suppression and glial cell line-derived neurotrophic factor gene expression in the hypothalamus of mice. Nutr Neurosci 2024; 27:106-119. [PMID: 36634108 DOI: 10.1080/1028415x.2023.2165938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The hypothalamus controls food intake by integrating nutrient signals, of which one of the most important is glucose. Consequently, impairments in hypothalamic glucose-sensing mechanisms are associated with hyperphagia and obesity. Environmental enrichment (EE) is an animal housing protocol that provides complex sensory, motor, and social stimulations and has been proven to reduce adiposity in laboratory mice. However, the mechanism by which EE promotes adiposity-suppressing effect remains incompletely understood. Neurotrophic factors play an important role in the development and maintenance of the nervous system, but they are also involved in the hypothalamic regulation of feeding. Brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF) are expressed in the hypothalamus and their expression is stimulated by glucose. EE is associated with increased expression of Bdnf mRNA in the hypothalamus. Therefore, we hypothesized that EE potentiates the anorectic action of glucose by altering the expression of neurotrophic factor genes in the hypothalamus. Male C57BL/6 mice were maintained under standard or EE conditions to investigate the feeding response to glucose and the associated expression of feeding-related neurotrophic factor genes in the hypothalamus. Intraperitoneal glucose injection reduced food intake in both control and EE mice with a significantly greater reduction in the EE group compared to the control group. EE caused a significantly enhanced response of Gdnf mRNA expression to glucose without altering basal Gdnf mRNA expression and Bdnf mRNA response to glucose. These findings suggest that EE enhances glucose-induced feeding suppression, at least partly, by enhancing hypothalamic glucose-sensing ability that involves GDNF.
Collapse
Affiliation(s)
- Muhammad S Aldhshan
- Division of Endocrinology and Metabolic Diseases, Department of Physiology & Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Tooru M Mizuno
- Division of Endocrinology and Metabolic Diseases, Department of Physiology & Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
4
|
Yang C, Mwangi SM, Balasubramaniam A, Li G, Merlin-Zhang O, Liu Y, Srinivasan S. Treatment of Obesity Through Glial Cell-Derived Neurotrophic Factor Lipid Nanoparticle Delivery in Mice. GASTRO HEP ADVANCES 2023; 3:38-47. [PMID: 38390283 PMCID: PMC10883424 DOI: 10.1016/j.gastha.2023.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
BACKGROUND AND AIMS The overexpression of glial cell-derived neurotrophic factor (GDNF) in the liver and adipose tissues offers strong protection against high-fat diet (HFD)-induced obesity in mice. We hypothesize that sustainably enhancing GDNF expression in the liver may provide a therapeutic effect that can prevent the progression of HFD-induced obesity in mice. METHODS Expression lentivector encoding mouse GDNF (GDNF(pDNA) or empty vector (pDNA, control) were encapsulated in lipid nanoparticles (LNPs) using the thin-film hydration method. Mice were fed with regular diet (RD) or HFD for 20 weeks prior to injection and the GDNF and control vector-loaded LNPs were administered by intravenous (IV) injection to mice once weekly for 5 weeks. Changes in body weight were monitored and mice tissues were collected and imaged for fluorescence using an IVIS in vivo imaging system. Post-treatment abdominal fat weight, colon length, and spleen weight were obtained. GDNF protein levels in the liver and serum were quantified by enzyme-linked immunosorbent assay, while liver AKT serine/threonine kinase and AMP-activated protein kinase phosphorylation levels were evaluated by Western blotting. RESULTS IV-injected GDNF(pDNA)-loaded LNPs targeted the liver and remained in there for up to 15 days postinjection. A single injection of GDNF(pDNA)-loaded LNPs significantly increased GDNF expression for 7 days and consequently increased the levels of phosphorylated AKT serine/threonine kinase and AMP-activated protein kinase. Once weekly injections of GDNF(pDNA)-loaded LNPs for 5 weeks slowed increase in body weight, reduced abdominal fat, and modulated the gut microbiota toward a healthier composition in HFD-fed mice. CONCLUSION GDNF(pDNA)-loaded LNPs could potentially be developed as a therapeutic strategy to reverse weight gain in obese patients.
Collapse
Affiliation(s)
- Chunhua Yang
- Institute for Biomedical Sciences, Center for Inflammation, Immunity & Infection, Digestive Disease Research Group, Georgia State University, Atlanta, Georgia
- Department of Research-Gastroenterology, Atlanta Veterans Affairs Health Care System, Decatur, Georgia
| | - Simon Musyoka Mwangi
- Department of Research-Gastroenterology, Atlanta Veterans Affairs Health Care System, Decatur, Georgia
- Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia
| | - Arun Balasubramaniam
- Department of Research-Gastroenterology, Atlanta Veterans Affairs Health Care System, Decatur, Georgia
- Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia
| | - Ge Li
- Department of Research-Gastroenterology, Atlanta Veterans Affairs Health Care System, Decatur, Georgia
- Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia
| | | | - Yunshan Liu
- Department of Research-Gastroenterology, Atlanta Veterans Affairs Health Care System, Decatur, Georgia
- Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia
| | - Shanthi Srinivasan
- Department of Research-Gastroenterology, Atlanta Veterans Affairs Health Care System, Decatur, Georgia
- Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
5
|
Ford MM, George BE, Van Laar VS, Holleran KM, Naidoo J, Hadaczek P, Vanderhooft LE, Peck EG, Dawes MH, Ohno K, Bringas J, McBride JL, Samaranch L, Forsayeth JR, Jones SR, Grant KA, Bankiewicz KS. GDNF gene therapy for alcohol use disorder in male non-human primates. Nat Med 2023; 29:2030-2040. [PMID: 37580533 PMCID: PMC10602124 DOI: 10.1038/s41591-023-02463-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 06/15/2023] [Indexed: 08/16/2023]
Abstract
Alcohol use disorder (AUD) exacts enormous personal, social and economic costs globally. Return to alcohol use in treatment-seeking patients with AUD is common, engendered by a cycle of repeated abstinence-relapse episodes even with use of currently available pharmacotherapies. Repeated ethanol use induces dopaminergic signaling neuroadaptations in ventral tegmental area (VTA) neurons of the mesolimbic reward pathway, and sustained dysfunction of reward circuitry is associated with return to drinking behavior. We tested this hypothesis by infusing adeno-associated virus serotype 2 vector encoding human glial-derived neurotrophic factor (AAV2-hGDNF), a growth factor that enhances dopaminergic neuron function, into the VTA of four male rhesus monkeys, with another four receiving vehicle, following induction of chronic alcohol drinking. GDNF expression ablated the return to alcohol drinking behavior over a 12-month period of repeated abstinence-alcohol reintroduction challenges. This behavioral change was accompanied by neurophysiological modulations to dopamine signaling in the nucleus accumbens that countered the hypodopaminergic signaling state associated with chronic alcohol use, indicative of a therapeutic modulation of limbic circuits countering the effects of alcohol. These preclinical findings suggest gene therapy targeting relapse prevention may be a potential therapeutic strategy for AUD.
Collapse
Affiliation(s)
- Matthew M Ford
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
- Department of Psychology, Lewis & Clark College, Portland, OR, USA
| | - Brianna E George
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Victor S Van Laar
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Katherine M Holleran
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Jerusha Naidoo
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Piotr Hadaczek
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Lauren E Vanderhooft
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Emily G Peck
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Monica H Dawes
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Kousaku Ohno
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - John Bringas
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Jodi L McBride
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Lluis Samaranch
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - John R Forsayeth
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Sara R Jones
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA.
| | - Krystof S Bankiewicz
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA.
- Department of Neurological Surgery, University of California, San Francisco, CA, USA.
| |
Collapse
|
6
|
Olfat S, Mätlik K, Kopra JJ, Garton DR, Iivanainen VH, Bhattacharya D, Jakobsson J, Piepponen TP, Andressoo JO. Increased Physiological GDNF Levels Have No Effect on Dopamine Neuron Protection and Restoration in a Proteasome Inhibition Mouse Model of Parkinson's Disease. eNeuro 2023; 10:ENEURO.0097-22.2023. [PMID: 36690469 PMCID: PMC9910577 DOI: 10.1523/eneuro.0097-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease that comprises a range of motor and nonmotor symptoms. Glial cell line-derived neurotrophic factor (GDNF) promotes the survival of dopamine neurons in vitro and in vivo, and intracranial delivery of GDNF has been tested in six clinical trials for treating PD. However, clinical trials with ectopic GDNF have yielded variable results, which could in part result from abnormal expression site and levels caused by ectopic overexpression. Therefore, an important open question is whether an increase in endogenous GDNF expression could be potent in reversing PD progression. Here, we tested the therapeutic potential of endogenous GDNF using mice in which endogenous GDNF can be conditionally upregulated specifically in cells that express GDNF naturally (conditional GDNF hypermorphic mice; GdnfcHyper ). We analyzed the impact of endogenous GDNF upregulation in both neuroprotection and neurorestoration procedures, and for both motor and nonmotor symptoms in the proteasome inhibitor lactacystin (LC) model of PD. Our results showed that upregulation of endogenous GDNF in the adult striatum is not protective in LC-induced PD model in mice. Since age is the largest risk factor for PD, we also analyzed the effect of deletion of endogenous GDNF in aged Gdnf conditional knock-out mice. We found that GDNF deletion does not increase susceptibility to LC-induced damage. We conclude that endogenous GDNF does not impact the outcome in the LC-induced proteasome inhibition mouse model of Parkinson's disease.
Collapse
Affiliation(s)
- Soophie Olfat
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm 17177, Sweden
- Department of Pharmacology, Faculty of Medicine, Neuroscience Center & Helsinki Institute of Life Science, University of Helsinki, Helsinki 00290, Finland
| | - Kärt Mätlik
- Department of Pharmacology, Faculty of Medicine, Neuroscience Center & Helsinki Institute of Life Science, University of Helsinki, Helsinki 00290, Finland
| | - Jaakko J Kopra
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Daniel R Garton
- Department of Pharmacology, Faculty of Medicine, Neuroscience Center & Helsinki Institute of Life Science, University of Helsinki, Helsinki 00290, Finland
| | - Vilma H Iivanainen
- Department of Pharmacology, Faculty of Medicine, Neuroscience Center & Helsinki Institute of Life Science, University of Helsinki, Helsinki 00290, Finland
| | - Dipabarna Bhattacharya
- Department of Pharmacology, Faculty of Medicine, Neuroscience Center & Helsinki Institute of Life Science, University of Helsinki, Helsinki 00290, Finland
| | - Johan Jakobsson
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, Lund 221 84, Sweden
| | - T Petteri Piepponen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Jaan-Olle Andressoo
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm 17177, Sweden
- Department of Pharmacology, Faculty of Medicine, Neuroscience Center & Helsinki Institute of Life Science, University of Helsinki, Helsinki 00290, Finland
| |
Collapse
|
7
|
Aldhshan MS, Jhanji G, Poritsanos NJ, Mizuno TM. Glucose Stimulates Glial Cell Line-Derived Neurotrophic Factor Gene Expression in Microglia through a GLUT5-Independent Mechanism. Int J Mol Sci 2022; 23:ijms23137073. [PMID: 35806073 PMCID: PMC9266953 DOI: 10.3390/ijms23137073] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 06/20/2022] [Accepted: 06/24/2022] [Indexed: 01/27/2023] Open
Abstract
Feeding-regulating neurotrophic factors are expressed in both neurons and glial cells. However, nutritional regulation of anorexigenic glial cell line-derived neurotrophic factor (GDNF) and orexigenic mesencephalic astrocyte-derived neurotrophic factor (MANF) expression in specific cell types remains poorly understood. Hypothalamic glucose sensing plays a critical role in the regulation of food intake. It has been theorized that local glucose concentration modulates microglial activity partially via glucose transporter 5 (GLUT5). We hypothesized that an increased local glucose concentration stimulates GDNF expression while inhibiting MANF expression in the hypothalamus and microglia via GLUT5. The present study investigated the effect of glucose on Gdnf and Manf mRNA expression in the mouse hypothalamus and murine microglial cell line SIM-A9. Intracerebroventricular glucose treatment significantly increased Gdnf mRNA levels in the hypothalamus without altering Manf mRNA levels. Exposure to high glucose caused a significant increase in Gdnf mRNA expression and a time-dependent change in Manf mRNA expression in SIM-A9 cells. GLUT5 inhibitor treatment did not block glucose-induced Gdnf mRNA expression in these cells. These findings suggest that microglia are responsive to changes in the local glucose concentration and increased local glucose availability stimulates the expression of microglial GNDF through a GLUT5-independent mechanism, contributing to glucose-induced feeding suppression.
Collapse
|
8
|
Liu X, Yang G, Sun T, Tao L, Shen D, Zhang W, Zhang J, Xue D, Chen B, Wu L, Liu C, Ma W. Glial cell line-derived neurotrophic factor contributes to alcoholic-induced liver injury by regulating the NF-κB pathway. Alcohol Clin Exp Res 2022; 46:724-735. [PMID: 35338490 DOI: 10.1111/acer.14815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 02/27/2022] [Accepted: 03/22/2022] [Indexed: 12/19/2022]
Abstract
BACKGROUND Alcoholic liver disease (ALD) is associated with high morbidity and mortality worldwide. The pathogenesis of ALD is not completely understood. Although accumulating evidence suggests an important role of glial cell line-derived neurotrophic factor (GDNF) in several diseases, there are no data concerning its role in ALD. This study compared patients with ALD with control subjects and used a mouse model and a cell culture model to investigate the function of GDNF in ALD and its mechanism of action in hepatocyte injury. METHODS Serum levels of GDNF were measured in 25 patients with ALD and 25 healthy control subjects. A 4-week Lieber-DeCarli ethanol (EtOH) liquid diet combined with the Gao-Binge model was used in the mouse study. Mouse primary hepatocytes and Huh-7 cells were used for cell experiments. The parameters of liver injury, inflammatory cytokines, and lipid metabolism were measured. RESULTS Patients with alcoholic hepatitis had higher serum GDNF than control subjects. Expression of GDNF mRNA and protein was markedly increased in mice in the chronic-plus-binge ALD mouse model. The level of GDNF mRNA was upregulated in primary hepatic stellate cells isolated from ethanol-fed mouse liver. Ethanol induced GDNF expression in LX2 cells. The levels of inflammatory cytokines (tumor necrosis factor α, interleukin 1β, and monocyte chemotactic protein 1) were significantly increased after GDNF stimulation in primary hepatocytes and Huh-7 cells. After GDNF stimulation, levels of both p-AKT and p-NF-κB were significantly increased in primary hepatocytes and Huh-7 cells. The NF-κB activity induced by GDNF was significantly decreased by an NF-κB inhibitor, which limited hepatocyte injury and inflammation. CONCLUSIONS The concentration of GDNF is increased in the circulation of ALD patients. GDNF promotes alcohol-induced liver injury and inflammation via the activation of NF-κB, which mediates hepatocyte injury and inflammatory cytokine expression. Based on these findings, GDNF is a potential therapeutic target for preventing or ameliorating liver injury in ALD.
Collapse
Affiliation(s)
- Xuling Liu
- Laboratory of Liver Disease, Department of Infectious Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guangyue Yang
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tiantian Sun
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Le Tao
- Laboratory of Liver Disease, Department of Infectious Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dongxiao Shen
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Zhang
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Zhang
- Laboratory of Liver Disease, Department of Infectious Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dongying Xue
- Laboratory of Liver Disease, Department of Infectious Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bei Chen
- Laboratory of Liver Disease, Department of Infectious Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liu Wu
- Laboratory of Liver Disease, Department of Infectious Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cheng Liu
- Laboratory of Liver Disease, Department of Infectious Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenting Ma
- Laboratory of Liver Disease, Department of Infectious Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
9
|
Elevated endogenous GDNF induces altered dopamine signalling in mice and correlates with clinical severity in schizophrenia. Mol Psychiatry 2022; 27:3247-3261. [PMID: 35618883 PMCID: PMC9708553 DOI: 10.1038/s41380-022-01554-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 11/08/2022]
Abstract
Presynaptic increase in striatal dopamine is the primary dopaminergic abnormality in schizophrenia, but the underlying mechanisms are not understood. Here, we hypothesized that increased expression of endogenous GDNF could induce dopaminergic abnormalities that resemble those seen in schizophrenia. To test the impact of GDNF elevation, without inducing adverse effects caused by ectopic overexpression, we developed a novel in vivo approach to conditionally increase endogenous GDNF expression. We found that a 2-3-fold increase in endogenous GDNF in the brain was sufficient to induce molecular, cellular, and functional changes in dopamine signalling in the striatum and prefrontal cortex, including increased striatal presynaptic dopamine levels and reduction of dopamine in prefrontal cortex. Mechanistically, we identified adenosine A2a receptor (A2AR), a G-protein coupled receptor that modulates dopaminergic signalling, as a possible mediator of GDNF-driven dopaminergic abnormalities. We further showed that pharmacological inhibition of A2AR with istradefylline partially normalised striatal GDNF and striatal and cortical dopamine levels in mice. Lastly, we found that GDNF levels are increased in the cerebrospinal fluid of first episode psychosis patients, and in post-mortem striatum of schizophrenia patients. Our results reveal a possible contributor for increased striatal dopamine signalling in a subgroup of schizophrenia patients and suggest that GDNF-A2AR crosstalk may regulate dopamine function in a therapeutically targetable manner.
Collapse
|
10
|
Klevebro S, Björkander S, Ekström S, Merid SK, Gruzieva O, Mälarstig A, Johansson Å, Kull I, Bergström A, Melén E. Inflammation-related plasma protein levels and association with adiposity measurements in young adults. Sci Rep 2021; 11:11391. [PMID: 34059769 PMCID: PMC8166979 DOI: 10.1038/s41598-021-90843-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/07/2021] [Indexed: 02/08/2023] Open
Abstract
Obesity-related inflammation is associated with cardiovascular, metabolic, and pulmonary diseases. The aim of this study was to demonstrate associations between adiposity measurements and levels of inflammation-related plasma proteins in a population of young adults. Subjects from a population-based birth cohort with a mean age of 22.5 years were included in the study population (n = 2074). Protein levels were analyzed using the Olink Proseek Multiplex Inflammation panel. Percentage body fat (%BF) and visceral fat rating (VFR) measurements were collected using Tanita MC 780 body composition monitor. Linear regression of standardized values was used to investigate associations. Potential effect modifications by sex and BMI category were assessed. Of 71 investigated proteins, 54 were significantly associated with all adiposity measurements [%BF, body mass index (BMI), VFR and waist circumference]. Among proteins associated with %BF, seven showed a larger or unique association in overweight/obese subjects and three showed a significant effect modification by sex. Fourteen proteins more strongly associated with VFR in females compared to males. Adipose-associated systemic inflammation was observed in this young adult population. Sex and adiposity localization influenced some of the associations. Our results highlight specific proteins as suitable biomarkers related to adiposity.
Collapse
Affiliation(s)
- Susanna Klevebro
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Sjukhusbacken 10, 118 83, Stockholm, Sweden.
- Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden.
| | - Sophia Björkander
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Sjukhusbacken 10, 118 83, Stockholm, Sweden
| | - Sandra Ekström
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Centre for Occupational and Environmental Medicine, Region Stockholm, Stockholm, Sweden
| | - Simon K Merid
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Sjukhusbacken 10, 118 83, Stockholm, Sweden
| | - Olena Gruzieva
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Centre for Occupational and Environmental Medicine, Region Stockholm, Stockholm, Sweden
| | - Anders Mälarstig
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Åsa Johansson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Inger Kull
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Sjukhusbacken 10, 118 83, Stockholm, Sweden
- Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Anna Bergström
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Centre for Occupational and Environmental Medicine, Region Stockholm, Stockholm, Sweden
| | - Erik Melén
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Sjukhusbacken 10, 118 83, Stockholm, Sweden
- Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| |
Collapse
|
11
|
Ingles-Prieto A, Furthmann N, Crossman SH, Tichy AM, Hoyer N, Petersen M, Zheden V, Biebl J, Reichhart E, Gyoergy A, Siekhaus DE, Soba P, Winklhofer KF, Janovjak H. Optogenetic delivery of trophic signals in a genetic model of Parkinson's disease. PLoS Genet 2021; 17:e1009479. [PMID: 33857132 PMCID: PMC8049241 DOI: 10.1371/journal.pgen.1009479] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/10/2021] [Indexed: 12/19/2022] Open
Abstract
Optogenetics has been harnessed to shed new mechanistic light on current and future therapeutic strategies. This has been to date achieved by the regulation of ion flow and electrical signals in neuronal cells and neural circuits that are known to be affected by disease. In contrast, the optogenetic delivery of trophic biochemical signals, which support cell survival and are implicated in degenerative disorders, has never been demonstrated in an animal model of disease. Here, we reengineered the human and Drosophila melanogaster REarranged during Transfection (hRET and dRET) receptors to be activated by light, creating one-component optogenetic tools termed Opto-hRET and Opto-dRET. Upon blue light stimulation, these receptors robustly induced the MAPK/ERK proliferative signaling pathway in cultured cells. In PINK1B9 flies that exhibit loss of PTEN-induced putative kinase 1 (PINK1), a kinase associated with familial Parkinson's disease (PD), light activation of Opto-dRET suppressed mitochondrial defects, tissue degeneration and behavioral deficits. In human cells with PINK1 loss-of-function, mitochondrial fragmentation was rescued using Opto-dRET via the PI3K/NF-кB pathway. Our results demonstrate that a light-activated receptor can ameliorate disease hallmarks in a genetic model of PD. The optogenetic delivery of trophic signals is cell type-specific and reversible and thus has the potential to inspire novel strategies towards a spatio-temporal regulation of tissue repair.
Collapse
Affiliation(s)
- Alvaro Ingles-Prieto
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Nikolas Furthmann
- Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Samuel H. Crossman
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, Australia
| | - Alexandra-Madelaine Tichy
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, Australia
| | - Nina Hoyer
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Meike Petersen
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa Zheden
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Julia Biebl
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Eva Reichhart
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, Australia
| | - Attila Gyoergy
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Daria E. Siekhaus
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Peter Soba
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Konstanze F. Winklhofer
- Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Harald Janovjak
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, Australia
- * E-mail:
| |
Collapse
|
12
|
Kambey PA, Kanwore K, Ayanlaja AA, Nadeem I, Du Y, Buberwa W, Liu W, Gao D. Failure of Glial Cell-Line Derived Neurotrophic Factor (GDNF) in Clinical Trials Orchestrated By Reduced NR4A2 (NURR1) Transcription Factor in Parkinson's Disease. A Systematic Review. Front Aging Neurosci 2021; 13:645583. [PMID: 33716718 PMCID: PMC7943926 DOI: 10.3389/fnagi.2021.645583] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/29/2021] [Indexed: 12/23/2022] Open
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative maladies with unforeseen complex pathologies. While this neurodegenerative disorder’s neuropathology is reasonably well known, its etiology remains a mystery, making it challenging to aim therapy. Glial cell-line derived neurotrophic factor (GDNF) remains an auspicious therapeutic molecule for treating PD. Neurotrophic factor derived from glial cell lines is effective in rodents and nonhuman primates, but clinical findings have been equivocal. Laborious exertions have been made over the past few decades to improve and assess GDNF in treating PD (clinical studies). Definitive clinical trials have, however, failed to demonstrate a survival advantage. Consequently, there seemed to be a doubt as to whether GDNF has merit in the potential treatment of PD. The purpose of this cutting edge review is to speculate as to why the clinical trials have failed to meet the primary endpoint. We introduce a hypothesis, “Failure of GDNF in clinical trials succumbed by nuclear receptor-related factor 1 (Nurr1) shortfall.” We demonstrate how Nurr1 binds to GDNF to induce dopaminergic neuron synthesis. Due to its undisputable neuro-protection aptitude, we display Nurr1 (also called Nr4a2) as a promising therapeutic target for PD.
Collapse
Affiliation(s)
- Piniel Alphayo Kambey
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Kouminin Kanwore
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Abiola Abdulrahman Ayanlaja
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Iqra Nadeem
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - YinZhen Du
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | | | - WenYa Liu
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Dianshuai Gao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
13
|
Terse PS, Kells AP, Noker P, Wright JF, Bankiewicz KS. Safety Assessment of AAV2-hGDNF Administered Via Intracerebral Injection in Rats for Treatment of Parkinson's Disease. Int J Toxicol 2021; 40:4-14. [PMID: 33131343 PMCID: PMC8171122 DOI: 10.1177/1091581820966315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is a potent neuroprotective biologic in Parkinson's disease models. Adeno-associated viral vector serotype 2 (AAV2)-human GDNF safety was assessed in rats treated with a single intracerebral dose of vehicle, 6.8 × 108, 6.8 × 109, or 5.2 × 1010 vector genomes (vg)/dose followed by interim sacrifices on day 7, 31, 90, and 376. There were no treatment-related effects observed on food consumption, body weight, hematology, clinical chemistry, coagulation parameters, neurobehavioral parameters, organ weights, or serum GDNF and anti-GDNF antibody levels. Increased serum anti-AAV2 neutralizing antibody titers were observed in the 5.2 × 1010 vg/dose group. Histopathological lesions were observed at the injection site in the 6.8 × 109 vg/dose (day 7) and 5.2 × 1010 vg/dose groups (days 7 and 31) and consisted of gliosis, mononuclear perivascular cuffing, intranuclear inclusion bodies, and/or apoptosis on day 7 and mononuclear perivascular cuffing on day 31. GDNF immunostaining was observed in the injection site in all dose groups through day 376 indicating no detectable impacts of anti-AAV2 neutralizing antibody. There was no evidence of increased expression of calcitonin gene-related peptide or Swann cell hyperplasia in the cervical and lumbar spinal cord or medulla oblongata at the 5.2 × 1010 vg/dose level indicating lack of hyperplastic effects. In conclusion, no systemic toxicity was observed, and the local toxicity observed at the injection site appeared to be reversible demonstrating a promising safety profile of intracerebral AAV2-GDNF delivery. Furthermore, an intracerebral dose of 6.8 × 108 AAV2-GDNF vg/dose was considered to be a no observed adverse effect level in rats.
Collapse
Affiliation(s)
- Pramod S. Terse
- National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | | | | | - J Fraser Wright
- Center for Definitive and Curative Medicine, Stanford University School of Medicine, CA, USA
| | | |
Collapse
|
14
|
Manfredsson FP, Polinski NK, Subramanian T, Boulis N, Wakeman DR, Mandel RJ. The Future of GDNF in Parkinson's Disease. Front Aging Neurosci 2020; 12:593572. [PMID: 33364933 PMCID: PMC7750181 DOI: 10.3389/fnagi.2020.593572] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Affiliation(s)
- Fredric P Manfredsson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Nicole K Polinski
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, United States
| | - Thyagarajan Subramanian
- Department of Neurology and Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, PA, United States
| | - Nicholas Boulis
- Department of Neurosurgery, Emory University, Atlanta, GA, United States
| | - Dustin R Wakeman
- Virscio, Inc., New Haven, CT, United States.,Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States
| | - Ronald J Mandel
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
| |
Collapse
|
15
|
Small Molecules and Peptides Targeting Glial Cell Line-Derived Neurotrophic Factor Receptors for the Treatment of Neurodegeneration. Int J Mol Sci 2020; 21:ijms21186575. [PMID: 32911810 PMCID: PMC7554781 DOI: 10.3390/ijms21186575] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/31/2020] [Accepted: 09/06/2020] [Indexed: 12/14/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) family ligands (GFLs) are able to promote the survival of multiple neuronal populations in the body and, therefore, hold considerable promise for disease-modifying treatments of diseases and conditions caused by neurodegeneration. Available data reveal the potential of GFLs for the therapy of Parkinson's disease, neuropathic pain and diseases caused by retinal degeneration but, also, amyotrophic lateral sclerosis and, possibly, Alzheimer's disease. Despite promising data collected in preclinical models, clinical translation of GFLs is yet to be conducted. The main reasons for the limited success of GFLs clinical development are the poor pharmacological characteristics of GFL proteins, such as the inability of GFLs to cross tissue barriers, poor diffusion in tissues, biphasic dose-response and activation of several receptors in the organism in different cell types, along with ethical limitations on patients' selection in clinical trials. The development of small molecules selectively targeting particular GFL receptors with improved pharmacokinetic properties can overcome many of the difficulties and limitations associated with the clinical use of GFL proteins. The current review lists several strategies to target the GFL receptor complex with drug-like molecules, discusses their advantages, provides an overview of available chemical scaffolds and peptides able to activate GFL receptors and describes the effects of these molecules in cultured cells and animal models.
Collapse
|
16
|
Nilsson IAK, Millischer V, Göteson A, Hübel C, Thornton LM, Bulik CM, Schalling M, Landén M. Aberrant inflammatory profile in acute but not recovered anorexia nervosa. Brain Behav Immun 2020; 88:718-724. [PMID: 32389698 DOI: 10.1016/j.bbi.2020.05.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 02/07/2023] Open
Abstract
Anorexia nervosa (AN) is a severe psychiatric disorder with high mortality and relapse rates. Even though changes in inflammatory markers and cytokines are known to accompany cachexia associated with somatic disorders such as cancer and chronic kidney disorder, studies on inflammatory markers in AN are rare and typically include few individuals. Here, we utilize an Olink Proteomics inflammatory panel to explore the concentrations of 92 preselected inflammation-related proteins in plasma samples from women with active AN (N = 113), recovered from AN (AN-REC, N = 113), and normal weight healthy controls (N = 114). After correction for multiple testing, twenty-five proteins differed significantly between the AN group and controls (lower levels: ADA, CCL19, CD40, CD5, CD8A, CSF1, CXCL1, CXCL5, HGF, IL10RB, IL12B, 1L18R1, LAP TGFß1, MCP3, OSM, TGFα, TNFRSF9, TNFS14 and TRANCE; higher levels: CCL11, CCL25, CST5, DNER, LIFR and OPG). Although more than half of these differences (N = 15) were present in the comparison between AN and AN-REC, no significant differences were seen between AN-REC and controls. Furthermore, twenty-five proteins correlated positively with BMI (ADA, AXIN1, CASP8, CD5, CD40, CSF1, CXCL1, CXCL5, EN-RAGE, HGF, IL6, IL10RB, IL12B, IL18, IL18R1, LAP TGFß1, OSM, SIRT2, STAMBP, TGFα, TNFRSF9, TNFS14, TRANCE, TRAIL and VEGFA) and four proteins correlated negatively with BMI (CCL11, CCL25, CCL28 and DNER). These results suggest that a dysregulated inflammatory status is associated with AN, but, importantly, seem to be confined to the acute illness state.
Collapse
Affiliation(s)
- Ida A K Nilsson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Centre for Eating Disorders Innovation, Karolinska Institutet, Stockholm, Sweden.
| | - Vincent Millischer
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Andreas Göteson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Christopher Hübel
- Centre for Eating Disorders Innovation, Karolinska Institutet, Stockholm, Sweden; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; UK National Institute for Health Research (NIHR) Biomedical Research Centre, South London and Maudsley Hospital, London, UK
| | - Laura M Thornton
- Department of Psychiatry, University of North Carolina at Chapel Hill, NC, USA
| | - Cynthia M Bulik
- Centre for Eating Disorders Innovation, Karolinska Institutet, Stockholm, Sweden; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Department of Psychiatry, University of North Carolina at Chapel Hill, NC, USA; Department of Nutrition, University of North Carolina at Chapel Hill, NC, USA
| | - Martin Schalling
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Mikael Landén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
17
|
Grassi S, Giussani P, Mauri L, Prioni S, Sonnino S, Prinetti A. Lipid rafts and neurodegeneration: structural and functional roles in physiologic aging and neurodegenerative diseases. J Lipid Res 2020; 61:636-654. [PMID: 31871065 PMCID: PMC7193971 DOI: 10.1194/jlr.tr119000427] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/11/2019] [Indexed: 12/14/2022] Open
Abstract
Lipid rafts are small, dynamic membrane areas characterized by the clustering of selected membrane lipids as the result of the spontaneous separation of glycolipids, sphingolipids, and cholesterol in a liquid-ordered phase. The exact dynamics underlying phase separation of membrane lipids in the complex biological membranes are still not fully understood. Nevertheless, alterations in the membrane lipid composition affect the lateral organization of molecules belonging to lipid rafts. Neural lipid rafts are found in brain cells, including neurons, astrocytes, and microglia, and are characterized by a high enrichment of specific lipids depending on the cell type. These lipid rafts seem to organize and determine the function of multiprotein complexes involved in several aspects of signal transduction, thus regulating the homeostasis of the brain. The progressive decline of brain performance along with physiological aging is at least in part associated with alterations in the composition and structure of neural lipid rafts. In addition, neurodegenerative conditions, such as lysosomal storage disorders, multiple sclerosis, and Parkinson's, Huntington's, and Alzheimer's diseases, are frequently characterized by dysregulated lipid metabolism, which in turn affects the structure of lipid rafts. Several events underlying the pathogenesis of these diseases appear to depend on the altered composition of lipid rafts. Thus, the structure and function of lipid rafts play a central role in the pathogenesis of many common neurodegenerative diseases.jlr;61/5/636/F1F1f1.
Collapse
Affiliation(s)
- Sara Grassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Paola Giussani
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Laura Mauri
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Simona Prioni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Sandro Sonnino
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Alessandro Prinetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy. mailto:
| |
Collapse
|
18
|
Turconi G, Kopra J, Võikar V, Kulesskaya N, Vilenius C, Piepponen TP, Andressoo JO. Chronic 2-Fold Elevation of Endogenous GDNF Levels Is Safe and Enhances Motor and Dopaminergic Function in Aged Mice. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:831-842. [PMID: 32368564 PMCID: PMC7191127 DOI: 10.1016/j.omtm.2020.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/02/2020] [Indexed: 02/08/2023]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) supports function and survival of dopamine neurons that degenerate in Parkinson’s disease (PD). Ectopic delivery of GDNF in clinical trials to treat PD is safe but lacks significant therapeutic effect. In pre-clinical models, ectopic GDNF is effective but causes adverse effects, including downregulation of tyrosine hydroxylase, only a transient boost in dopamine metabolism, aberrant neuronal sprouting, and hyperactivity. Hindering development of GDNF mimetic increased signaling via GDNF receptor RET by activating mutations results in cancer. Safe and effective mode of action must be defined first in animal models to develop successful GDNF-based therapies. Previously we showed that about a 2-fold increase in endogenous GDNF expression is safe and results in increased motor and dopaminergic function and protection in a PD model in young animals. Recently, similar results were reported using a novel Gdnf mRNA-targeting strategy. Next, it is important to establish the safety of a long-term increase in endogenous GDNF expression. We report behavioral, dopamine system, and cancer analysis of five cohorts of aged mice with a 2-fold increase in endogenous GDNF. We found a sustained increase in dopamine levels, improvement in motor learning, and no side effects or cancer. These results support the rationale for further development of endogenous GDNF-based treatments and GDNF mimetic.
Collapse
Affiliation(s)
- Giorgio Turconi
- Department of Pharmacology, Faculty of Medicine and Helsinki Institute of Life Science, Haartmaninkatu 8, University of Helsinki, Helsinki 00014, Finland
| | - Jaakko Kopra
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, Viikinkaari 5E, University of Helsinki, Helsinki 00014, Finland
| | - Vootele Võikar
- Neuroscience Center/Laboratory Animal Center, Mustialankatu 1, University of Helsinki, Helsinki 00014, Finland
| | - Natalia Kulesskaya
- Neuroscience Center/Laboratory Animal Center, Mustialankatu 1, University of Helsinki, Helsinki 00014, Finland
| | - Carolina Vilenius
- Institute of Biotechnology, Viikinkaari 5D, University of Helsinki, Helsinki 00014, Finland
| | - T Petteri Piepponen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, Viikinkaari 5E, University of Helsinki, Helsinki 00014, Finland
| | - Jaan-Olle Andressoo
- Department of Pharmacology, Faculty of Medicine and Helsinki Institute of Life Science, Haartmaninkatu 8, University of Helsinki, Helsinki 00014, Finland.,Institute of Biotechnology, Viikinkaari 5D, University of Helsinki, Helsinki 00014, Finland.,Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm 141 83, Sweden
| |
Collapse
|
19
|
Bao XQ, Wang L, Yang HY, Hou LY, Wang QS, Zhang D. Induction of glial cell line-derived neurotrophic factor by the squamosamide derivative FLZ in astroglia has neuroprotective effects on dopaminergic neurons. Brain Res Bull 2019; 154:32-42. [PMID: 31669104 DOI: 10.1016/j.brainresbull.2019.10.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 10/08/2019] [Accepted: 10/19/2019] [Indexed: 12/27/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) has neurotrophic activity for the survival of dopaminergic neurons, which is under active investigation for Parkinson's disease (PD) therapy. FLZ is a potential new drug for PD treatment. However, it is unclear whether neurotrophic activity contributes to the neuroprotective effects of FLZ. Here we found that FLZ markedly improved the function of dopaminergic neurons in primary mesencephalic neuron/glia cultures. Further investigation demonstrated that astroglia were required for FLZ to function as a neurotrophic regulator, as FLZ failed to show neurotrophic effects in the absence of astroglia. We clarified that GDNF was responsible for the neurotrophic effects of FLZ since FLZ selectively stimulated GDNF production, which was confirmed by the finding that the neurotrophic effect of FLZ was attenuated by GDNF-neutralizing antibody. Mechanistic study demonstrated that GDNF induction by FLZ was CREB-dependent and that PI3K/Akt was the main pathway regulating CREB activity, which was confirmed by in vivo studies. We also validated that the induction of GDNF by FLZ contributed to PD treatment in vivo. In conclusion, the present data provided evidence that FLZ had robust neurotrophic effects on dopaminergic neurons through sustained induction of GDNF in astroglia by activating the PI3K/Akt/CREB pathway.
Collapse
Affiliation(s)
- Xiu-Qi Bao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College Xian Nong Tan Street, Beijing, 100050, China
| | - Lu Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College Xian Nong Tan Street, Beijing, 100050, China
| | - Han-Yu Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College Xian Nong Tan Street, Beijing, 100050, China
| | - Li-Yan Hou
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
| | - Qing-Shan Wang
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China.
| | - Dan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College Xian Nong Tan Street, Beijing, 100050, China.
| |
Collapse
|
20
|
Burnside ER, De Winter F, Didangelos A, James ND, Andreica EC, Layard-Horsfall H, Muir EM, Verhaagen J, Bradbury EJ. Immune-evasive gene switch enables regulated delivery of chondroitinase after spinal cord injury. Brain 2019; 141:2362-2381. [PMID: 29912283 PMCID: PMC6061881 DOI: 10.1093/brain/awy158] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 04/22/2018] [Indexed: 12/12/2022] Open
Abstract
Chondroitinase ABC is a promising preclinical therapy that promotes functional neuroplasticity after CNS injury by degrading extracellular matrix inhibitors. Efficient delivery of chondroitinase ABC to the injured mammalian spinal cord can be achieved by viral vector transgene delivery. This approach dramatically modulates injury pathology and restores sensorimotor functions. However, clinical development of this therapy is limited by a lack of ability to exert control over chondroitinase gene expression. Prior experimental gene regulation platforms are likely to be incompatible with the non-resolving adaptive immune response known to occur following spinal cord injury. Therefore, here we apply a novel immune-evasive dual vector system, in which the chondroitinase gene is under a doxycycline inducible regulatory switch, utilizing a chimeric transactivator designed to evade T cell recognition. Using this novel vector system, we demonstrate tight temporal control of chondroitinase ABC gene expression, effectively removing treatment upon removal of doxycycline. This enables a comparison of short and long-term gene therapy paradigms in the treatment of clinically-relevant cervical level contusion injuries in adult rats. We reveal that transient treatment (2.5 weeks) is sufficient to promote improvement in sensory axon conduction and ladder walking performance. However, in tasks requiring skilled reaching and grasping, only long term treatment (8 weeks) leads to significantly improved function, with rats able to accurately grasp and retrieve sugar pellets. The late emergence of skilled hand function indicates enhanced neuroplasticity and connectivity and correlates with increased density of vGlut1+ innervation in spinal cord grey matter, particularly in lamina III–IV above and below the injury. Thus, our novel gene therapy system provides an experimental tool to study temporal effects of extracellular matrix digestion as well as an encouraging step towards generating a safer chondroitinase gene therapy strategy, longer term administration of which increases neuroplasticity and recovery of descending motor control. This preclinical study could have a significant impact for tetraplegic individuals, for whom recovery of hand function is an important determinant of independence, and supports the ongoing development of chondroitinase gene therapy towards clinical application for the treatment of spinal cord injury.
Collapse
Affiliation(s)
- Emily R Burnside
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), Guy's Campus, London Bridge, London, SE1 1UL, UK
| | - Fred De Winter
- Netherlands Institute for Neuroscience, Laboratory for Neuroregeneration, 1105 BA Amsterdam, The Netherlands
| | - Athanasios Didangelos
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), Guy's Campus, London Bridge, London, SE1 1UL, UK
| | - Nicholas D James
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), Guy's Campus, London Bridge, London, SE1 1UL, UK
| | - Elena-Cristina Andreica
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), Guy's Campus, London Bridge, London, SE1 1UL, UK
| | - Hugo Layard-Horsfall
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), Guy's Campus, London Bridge, London, SE1 1UL, UK
| | - Elizabeth M Muir
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB2 3EG, UK
| | - Joost Verhaagen
- Netherlands Institute for Neuroscience, Laboratory for Neuroregeneration, 1105 BA Amsterdam, The Netherlands
| | - Elizabeth J Bradbury
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), Guy's Campus, London Bridge, London, SE1 1UL, UK
| |
Collapse
|
21
|
Cheng S, Tereshchenko J, Zimmer V, Vachey G, Pythoud C, Rey M, Liefhebber J, Raina A, Streit F, Mazur A, Bähr M, Konstantinova P, Déglon N, Kügler S. Therapeutic efficacy of regulable GDNF expression for Huntington's and Parkinson's disease by a high-induction, background-free “GeneSwitch” vector. Exp Neurol 2018; 309:79-90. [DOI: 10.1016/j.expneurol.2018.07.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 06/22/2018] [Accepted: 07/31/2018] [Indexed: 02/02/2023]
|
22
|
Mätlik K, Võikar V, Vilenius C, Kulesskaya N, Andressoo JO. Two-fold elevation of endogenous GDNF levels in mice improves motor coordination without causing side-effects. Sci Rep 2018; 8:11861. [PMID: 30089897 PMCID: PMC6082872 DOI: 10.1038/s41598-018-29988-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/20/2018] [Indexed: 01/11/2023] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) promotes the survival of dopaminergic neurons in vitro and in vivo. For this reason, GDNF is currently in clinical trials for the treatment of Parkinson’s disease (PD). However, how endogenous GDNF influences dopamine system function and animal behavior is not fully understood. We recently generated GDNF hypermorphic mice that express increased levels of endogenous GDNF from the native locus, resulting in augmented function of the nigrostriatal dopamine system. Specifically, Gdnf wt/hyper mice have a mild increase in striatal and midbrain dopamine levels, increased dopamine transporter activity, and 15% increased numbers of midbrain dopamine neurons and striatal dopaminergic varicosities. Since changes in the dopamine system are implicated in several neuropsychiatric diseases, including schizophrenia, attention deficit hyperactivity disorder (ADHD) and depression, and ectopic GDNF delivery associates with side-effects in PD models and clinical trials, we further investigated Gdnf wt/hyper mice using 20 behavioral tests. Despite increased dopamine levels, dopamine release and dopamine transporter activity, there were no differences in psychiatric disease related phenotypes. However, compared to controls, male Gdnf wt/hyper mice performed better in tests measuring motor function. Therefore, a modest elevation of endogenous GDNF levels improves motor function but does not induce adverse behavioral outcomes.
Collapse
Affiliation(s)
- Kärt Mätlik
- Department of Pharmacology, Faculty of Medicine & Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Vootele Võikar
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Carolina Vilenius
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Natalia Kulesskaya
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Jaan-Olle Andressoo
- Department of Pharmacology, Faculty of Medicine & Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland. .,Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
23
|
Intranasal Delivery of pGDNF DNA Nanoparticles Provides Neuroprotection in the Rat 6-Hydroxydopamine Model of Parkinson’s Disease. Mol Neurobiol 2018; 56:688-701. [DOI: 10.1007/s12035-018-1109-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 05/03/2018] [Indexed: 10/16/2022]
|
24
|
Das AT, Tenenbaum L, Berkhout B. Tet-On Systems For Doxycycline-inducible Gene Expression. Curr Gene Ther 2017; 16:156-67. [PMID: 27216914 PMCID: PMC5070417 DOI: 10.2174/1566523216666160524144041] [Citation(s) in RCA: 244] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 05/03/2016] [Accepted: 05/03/2016] [Indexed: 11/22/2022]
Abstract
The tetracycline-controlled Tet-Off and Tet-On gene expression systems are used to regulate the activity of genes in eukaryotic cells in diverse settings, varying from basic biological research to biotechnology and gene therapy applications. These systems are based on regulatory elements that control the activity of the tetracycline-resistance operon in bacteria. The Tet-Off system allows silencing of gene expression by administration of tetracycline (Tc) or tetracycline-derivatives like doxycycline (dox), whereas the Tet-On system allows activation of gene expression by dox. Since the initial design and construction of the original Tet-system, these bacterium-derived systems have been significantly improved for their function in eukaryotic cells. We here review how a dox-controlled HIV-1 variant was designed and used to greatly improve the activity and dox-sensitivity of the rtTA transcriptional activator component of the Tet-On system. These optimized rtTA variants require less dox for activation, which will reduce side effects and allow gene control in tissues where a relatively low dox level can be reached, such as the brain.
Collapse
Affiliation(s)
- Atze T Das
- Laboratory of Experimental Virology, Academic Medical Center, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.
| | | | | |
Collapse
|
25
|
Yurek D, Hasselrot U, Sesenoglu-Laird O, Padegimas L, Cooper M. Intracerebral injections of DNA nanoparticles encoding for a therapeutic gene provide partial neuroprotection in an animal model of neurodegeneration. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:2209-2217. [PMID: 28666950 DOI: 10.1016/j.nano.2017.06.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/02/2017] [Accepted: 06/14/2017] [Indexed: 11/18/2022]
Abstract
This study reports proof of concept for administering compacted DNA nanoparticles (DNPs) intracerebrally as a means to protect against neurotoxin-induced neurodegeneration of dopamine (DA) neurons. In this study we used DNPs that encoded for human glial cell line-derived neurotrophic factor (hGDNF); GDNF is a potent neurotrophic factor for DA neurons. Intracerebral injections of DNPs into the striatum and/or substantia nigra were performed 1 week before treatment with a neurotoxin. We observed that the number of surviving DA cells, the density of DA fiber terminals and recovery of motor function were greater in animals injected with GDNF-encoding DNPs than in control animals receiving DNPs encoding for an inert reporter gene. The results of these studies are one of the first to demonstrate that a non-viral, synthetic nanoparticle can be used to deliver therapeutic genes to cells in the brain as a means to protect cells against neurotoxin-induced neurodegeneration.
Collapse
Affiliation(s)
- David Yurek
- Department of Neurosurgery, University of Kentucky College of Medicine, Lexington, KY; University of Kentucky Nanobiotechnology Center, Lexington, KY.
| | - Ulla Hasselrot
- Department of Neurosurgery, University of Kentucky College of Medicine, Lexington, KY
| | | | | | - Mark Cooper
- Copernicus Therapeutics, Inc., Cleveland, OH
| |
Collapse
|
26
|
Kanaan NM, Sellnow RC, Boye SL, Coberly B, Bennett A, Agbandje-McKenna M, Sortwell CE, Hauswirth WW, Boye SE, Manfredsson FP. Rationally Engineered AAV Capsids Improve Transduction and Volumetric Spread in the CNS. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 8:184-197. [PMID: 28918020 PMCID: PMC5503098 DOI: 10.1016/j.omtn.2017.06.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 06/14/2017] [Accepted: 06/15/2017] [Indexed: 12/13/2022]
Abstract
Adeno-associated virus (AAV) is the most common vector for clinical gene therapy of the CNS. This popularity originates from a high safety record and the longevity of transgene expression in neurons. Nevertheless, clinical efficacy for CNS indications is lacking, and one reason for this is the relatively limited spread and transduction efficacy in large regions of the human brain. Using rationally designed modifications of the capsid, novel AAV capsids have been generated that improve intracellular processing and result in increased transgene expression. Here, we sought to improve AAV-mediated neuronal transduction to minimize the existing limitations of CNS gene therapy. We investigated the efficacy of CNS transduction using a variety of tyrosine and threonine capsid mutants based on AAV2, AAV5, and AAV8 capsids, as well as AAV2 mutants incapable of binding heparan sulfate (HS). We found that mutating several tyrosine residues on the AAV2 capsid significantly enhanced neuronal transduction in the striatum and hippocampus, and the ablation of HS binding also increased the volumetric spread of the vector. Interestingly, the analogous tyrosine substitutions on AAV5 and AAV8 capsids did not improve the efficacy of these serotypes. Our results demonstrate that the efficacy of CNS gene transfer can be significantly improved with minor changes to the AAV capsid and that the effect is serotype specific.
Collapse
Affiliation(s)
- Nicholas M Kanaan
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI 49503, USA; Mercy Health Saint Mary's, Grand Rapids, MI 49503, USA
| | - Rhyomi C Sellnow
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Sanford L Boye
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - Ben Coberly
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI 49503, USA; Neuroscience Program, Michigan State University, East Lansing, MI 48825, USA
| | - Antonette Bennett
- Department of Biochemistry and Molecular Biology, The McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mavis Agbandje-McKenna
- Department of Biochemistry and Molecular Biology, The McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Caryl E Sortwell
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI 49503, USA; Mercy Health Saint Mary's, Grand Rapids, MI 49503, USA
| | - William W Hauswirth
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - Shannon E Boye
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - Fredric P Manfredsson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI 49503, USA; Mercy Health Saint Mary's, Grand Rapids, MI 49503, USA.
| |
Collapse
|
27
|
Mead BP, Kim N, Miller GW, Hodges D, Mastorakos P, Klibanov AL, Mandell JW, Hirsh J, Suk JS, Hanes J, Price RJ. Novel Focused Ultrasound Gene Therapy Approach Noninvasively Restores Dopaminergic Neuron Function in a Rat Parkinson's Disease Model. NANO LETTERS 2017; 17:3533-3542. [PMID: 28511006 PMCID: PMC5539956 DOI: 10.1021/acs.nanolett.7b00616] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Therapies capable of decelerating, or perhaps even halting, neurodegeneration in Parkinson's disease (PD) remain elusive. Clinical trials of PD gene therapy testing the delivery of neurotrophic factors, such as the glial cell-line derived neurotrophic factor (GDNF), have been largely ineffective due to poor vector distribution throughout the diseased regions in the brain. In addition, current delivery strategies involve invasive procedures that obviate the inclusion of early stage patients who are most likely to benefit from GDNF-based gene therapy. Here, we introduce a two-pronged treatment strategy, composed of MR image-guided focused ultrasound (FUS) and brain-penetrating nanoparticles (BPN), that provides widespread but targeted GDNF transgene expression in the brain following systemic administration. MR image-guided FUS allows circulating gene vectors to partition into the brain tissue by noninvasive and transient opening of the blood-brain barrier (BBB) within the areas where FUS is applied. Once beyond the BBB, BPN provide widespread and uniform GDNF expression throughout the targeted brain tissue. After only a single treatment, our strategy led to therapeutically relevant levels of GDNF protein content in the FUS-targeted regions in the striatum of the 6-OHDA-induced rat model of PD, which lasted at least up to 10 weeks. Importantly, our strategy restored both dopamine levels and dopaminergic neuron density and reversed behavioral indicators of PD-associated motor dysfunction with no evidence of local or systemic toxicity. Our combinatorial approach overcomes limitations of current delivery strategies, thereby potentially providing a novel means to treat PD.
Collapse
Affiliation(s)
- Brian P. Mead
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Namho Kim
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - G. Wilson Miller
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, United States
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia 22908, United States
| | - David Hodges
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Panagiotis Mastorakos
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Alexander L. Klibanov
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, United States
- Cardiovascular Division, University of Virginia, Charlottesville, Virginia 22908, United States
| | - James W. Mandell
- Department of Pathology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Jay Hirsh
- Department of Biology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Jung Soo Suk
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Justin Hanes
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Richard J. Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, United States
| |
Collapse
|
28
|
Sarcopenic obesity or obese sarcopenia: A cross talk between age-associated adipose tissue and skeletal muscle inflammation as a main mechanism of the pathogenesis. Ageing Res Rev 2017; 35:200-221. [PMID: 27702700 DOI: 10.1016/j.arr.2016.09.008] [Citation(s) in RCA: 458] [Impact Index Per Article: 65.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/23/2016] [Accepted: 09/26/2016] [Indexed: 02/08/2023]
Abstract
Sarcopenia, an age-associated decline in skeletal muscle mass coupled with functional deterioration, may be exacerbated by obesity leading to higher disability, frailty, morbidity and mortality rates. In the combination of sarcopenia and obesity, the state called sarcopenic obesity (SOB), some key age- and obesity-mediated factors and pathways may aggravate sarcopenia. This review will analyze the mechanisms underlying the pathogenesis of SOB. In obese adipose tissue (AT), adipocytes undergo hypertrophy, hyperplasia and activation resulted in accumulation of pro-inflammatory macrophages and other immune cells as well as dysregulated production of various adipokines that together with senescent cells and the immune cell-released cytokines and chemokines create a local pro-inflammatory status. In addition, obese AT is characterized by excessive production and disturbed capacity to store lipids, which accumulate ectopically in skeletal muscle. These intramuscular lipids and their derivatives induce mitochondrial dysfunction characterized by impaired β-oxidation capacity and increased reactive oxygen species formation providing lipotoxic environment and insulin resistance as well as enhanced secretion of some pro-inflammatory myokines capable of inducing muscle dysfunction by auto/paracrine manner. In turn, by endocrine manner, these myokines may exacerbate AT inflammation and also support chronic low grade systemic inflammation (inflammaging), overall establishing a detrimental vicious circle maintaining AT and skeletal muscle inflammation, thus triggering and supporting SOB development. Under these circumstances, we believe that AT inflammation dominates over skeletal muscle inflammation. Thus, in essence, it redirects the vector of processes from "sarcopenia→obesity" to "obesity→sarcopenia". We therefore propose that this condition be defined as "obese sarcopenia", to reflect the direction of the pathological pathway.
Collapse
|
29
|
Tenenbaum L, Humbert-Claude M. Glial Cell Line-Derived Neurotrophic Factor Gene Delivery in Parkinson's Disease: A Delicate Balance between Neuroprotection, Trophic Effects, and Unwanted Compensatory Mechanisms. Front Neuroanat 2017; 11:29. [PMID: 28442998 PMCID: PMC5385337 DOI: 10.3389/fnana.2017.00029] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 03/23/2017] [Indexed: 12/14/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) and Neurturin (NRTN) bind to a receptor complex consisting of a member of the GDNF family receptor (GFR)-α and the Ret tyrosine kinase. Both factors were shown to protect nigro-striatal dopaminergic neurons and reduce motor symptoms when applied terminally in toxin-induced Parkinson's disease (PD) models. However, clinical trials based on intraputaminal GDNF protein administration or recombinant adeno-associated virus (rAAV)-mediated NRTN gene delivery have been disappointing. In this review, several factors that could have limited the clinical benefits are discussed. Retrograde transport of GDNF/NRTN to the dopaminergic neurons soma is thought to be necessary for NRTN/GFR-α/Ret signaling mediating the pro-survival effect. Therefore, the feasibility of treating advanced patients with neurotrophic factors is questioned by recent data showing that: (i) tyrosine hydroxylase-positive putaminal innervation has almost completely disappeared at 5 years post-diagnosis and (ii) in patients enrolled in the rAAV-NRTN trial more than 5 years post-diagnosis, NRTN was almost not transported to the substantia nigra pars compacta. In addition to its anti-apoptotic and neurotrophic properties, GDNF also interferes with dopamine homeostasis via time and dose-dependent effects such as: stimulation of dopamine neuron excitability, inhibition of dopamine transporter activity, tyrosine hydroxylase phosphorylation, and inhibition of tyrosine hydroxylase transcription. Depending on the delivery parameters, the net result of this intricate network of regulations could be either beneficial or deleterious. In conclusion, further unraveling of the mechanism of action of GDNF gene delivery in relevant animal models is still needed to optimize the clinical benefits of this new therapeutic approach. Recent developments in the design of regulated viral vectors will allow to finely adjust the GDNF dose and period of administration. Finally, new clinical studies in less advanced patients are warranted to evaluate the potential of AAV-mediated neurotrophic factors gene delivery in PD. These will be facilitated by the demonstration of the safety of rAAV administration into the human brain.
Collapse
Affiliation(s)
- Liliane Tenenbaum
- Laboratory of Cellular and Molecular Neurotherapies, Clinical Neuroscience Department, Center for Neuroscience Research, Lausanne University HospitalLausanne, Switzerland
| | - Marie Humbert-Claude
- Laboratory of Cellular and Molecular Neurotherapies, Clinical Neuroscience Department, Center for Neuroscience Research, Lausanne University HospitalLausanne, Switzerland
| |
Collapse
|
30
|
Thomsen GM, Alkaslasi M, Vit JP, Lawless G, Godoy M, Gowing G, Shelest O, Svendsen CN. Systemic injection of AAV9-GDNF provides modest functional improvements in the SOD1 G93A ALS rat but has adverse side effects. Gene Ther 2017; 24:245-252. [PMID: 28276446 PMCID: PMC5404206 DOI: 10.1038/gt.2017.9] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 01/06/2017] [Accepted: 01/10/2017] [Indexed: 12/11/2022]
Abstract
Injecting proteins into the central nervous system that stimulate neuronal growth can lead to beneficial effects in animal models of disease. In particular, glial cell line-derived neurotrophic factor (GDNF) has shown promise in animal and cell models of Parkinson's disease, Huntington's disease and amyotrophic lateral sclerosis (ALS). Here, systemic AAV9-GDNF was delivered via tail vein injections to young rats to determine whether this could be a safe and functional strategy to treat the SOD1G93A rat model of ALS and, therefore, translated to a therapy for ALS patients. We found that GDNF administration in this manner resulted in modest functional improvement, whereby grip strength was maintained for longer and the onset of forelimb paralysis was delayed compared to non-treated rats. This did not, however, translate into an extension in survival. In addition, ALS rats receiving GDNF exhibited slower weight gain, reduced activity levels and decreased working memory. Collectively, these results confirm that caution should be applied when applying growth factors such as GDNF systemically to multiple tissues.
Collapse
Affiliation(s)
- G M Thomsen
- The Board of Governor's Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - M Alkaslasi
- The Board of Governor's Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - J-P Vit
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Biobehavioral Research Core, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - G Lawless
- The Board of Governor's Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - M Godoy
- The Board of Governor's Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - G Gowing
- The Board of Governor's Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - O Shelest
- The Board of Governor's Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - C N Svendsen
- The Board of Governor's Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
31
|
Stott SRW, Hayat S, Carnwath T, Garas S, Sleeman JP, Barker RA. CD24 expression does not affect dopamine neuronal survival in a mouse model of Parkinson's disease. PLoS One 2017; 12:e0171748. [PMID: 28182766 PMCID: PMC5300212 DOI: 10.1371/journal.pone.0171748] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/25/2017] [Indexed: 12/26/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative condition that is characterised by the loss of specific populations of neurons in the brain. The mechanisms underlying this selective cell death are unknown but by using laser capture microdissection, the glycoprotein, CD24 has been identified as a potential marker of the populations of cells that are affected in PD. Using in situ hybridization and immunohistochemistry on sections of mouse brain, we confirmed that CD24 is robustly expressed by many of these subsets of cells. To determine if CD24 may have a functional role in PD, we modelled the dopamine cell loss of PD in Cd24 mutant mice using striatal delivery of the neurotoxin 6-OHDA. We found that Cd24 mutant mice have an anatomically normal dopamine system and that this glycoprotein does not modulate the lesion effects of 6-OHDA delivered into the striatum. We then undertook in situ hybridization studies on sections of human brain and found-as in the mouse brain-that CD24 is expressed by many of the subsets of the cells that are vulnerable in PD, but not those of the midbrain dopamine system. Finally, we sought to determine if CD24 is required for the neuroprotective effect of Glial cell-derived neurotrophic factor (GDNF) on the dopaminergic nigrostriatal pathway. Our results indicate that in the absence of CD24, there is a reduction in the protective effects of GDNF on the dopaminergic fibres in the striatum, but no difference in the survival of the cell bodies in the midbrain. While we found no obvious role for CD24 in the normal development and maintenance of the dopaminergic nigrostriatal system in mice, it may have a role in mediating the neuroprotective aspects of GDNF in this system.
Collapse
Affiliation(s)
- Simon R. W Stott
- John van Geest Centre for Brain Repair, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge, England
- * E-mail:
| | - Shaista Hayat
- John van Geest Centre for Brain Repair, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge, England
| | - Tom Carnwath
- John van Geest Centre for Brain Repair, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge, England
| | - Shaady Garas
- John van Geest Centre for Brain Repair, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge, England
| | - Jonathan P. Sleeman
- Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Roger A. Barker
- John van Geest Centre for Brain Repair, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge, England
- Wellcome Trust-MRC Stem Cell Institute, Cambridge, England
| |
Collapse
|
32
|
Ibáñez CF, Andressoo JO. Biology of GDNF and its receptors — Relevance for disorders of the central nervous system. Neurobiol Dis 2017; 97:80-89. [DOI: 10.1016/j.nbd.2016.01.021] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/14/2016] [Accepted: 01/25/2016] [Indexed: 01/15/2023] Open
|
33
|
Franco-Robles E, López MG. Agavins Increase Neurotrophic Factors and Decrease Oxidative Stress in the Brains of High-Fat Diet-Induced Obese Mice. Molecules 2016; 21:molecules21080998. [PMID: 27490526 PMCID: PMC6273709 DOI: 10.3390/molecules21080998] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 07/18/2016] [Accepted: 07/26/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Fructans obtained from agave, called agavins, have recently shown significant benefits for human health including obesity. Therefore, we evaluated the potential of agavins as neuroprotectors and antioxidants by determining their effect on brain-derived neurotrophic factor (BDNF) and glial-derived neurotrophic factor (GDNF) as well as oxidative brain damage in of obese mice. METHODS Male C57BL/6J mice were fed a high-fat diet (HFD) and treated daily with 5% (HFD/A5) or 10% (HFD/A10) of agavins or a standard diet (SD) for 10 weeks. The levels of BDNF and GDNF were evaluated by ELISA. The oxidative stress was evaluated by lipid peroxidation (TBARS) and carbonyls. SCFAs were also measured with GC-FID. Differences between groups were assessed using ANOVA and by Tukey's test considering p < 0.05. RESULTS The body weight gain and food intake of mice HFD/A10 group were significantly lower than those in the HFD group. Agavins restored BDNF levels in HFD/A5 group and GDNF levels of HFD/A5 and HFD/A10 groups in cerebellum. Interestingly, agavins decreased TBARS levels in HFD/A5 and HFD/A10 groups in the hippocampus, frontal cortex and cerebellum. Carbonyl levels were also lower in HFD/A5 and HFD/A10 for only the hippocampus and cerebellum. It was also found that agavins enhanced SCFAs production in feces. CONCLUSION Agavins may act as bioactive ingredients with antioxidant and protective roles in the brain.
Collapse
Affiliation(s)
- Elena Franco-Robles
- Departamento de Biotecnología y Bioquímica, Centro de Investigación y de Estudios Avanzados del IPN, Unidad de Biotecnología e Ingeniería Genética de Plantas, Irapuato, Guanajuato C.P. 36821, México.
| | - Mercedes G López
- Departamento de Biotecnología y Bioquímica, Centro de Investigación y de Estudios Avanzados del IPN, Unidad de Biotecnología e Ingeniería Genética de Plantas, Irapuato, Guanajuato C.P. 36821, México.
| |
Collapse
|
34
|
Marks WJ, Baumann TL, Bartus RT. Long-Term Safety of Patients with Parkinson's Disease Receiving rAAV2-Neurturin (CERE-120) Gene Transfer. Hum Gene Ther 2016; 27:522-7. [DOI: 10.1089/hum.2015.134] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- William J. Marks
- University of California, San Francisco, San Francisco, California
| | | | | | | |
Collapse
|
35
|
A regulatable AAV vector mediating GDNF biological effects at clinically-approved sub-antimicrobial doxycycline doses. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 5:16027. [PMID: 27069954 PMCID: PMC4813607 DOI: 10.1038/mtm.2016.27] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/26/2016] [Accepted: 02/26/2016] [Indexed: 12/19/2022]
Abstract
Preclinical and clinical data stress the importance of pharmacologically-controlling glial cell line-derived neurotrophic factor (GDNF) intracerebral administration to treat PD. The main challenge is finding a combination of a genetic switch and a drug which, when administered at a clinically-approved dose, reaches the brain in sufficient amounts to induce a therapeutic effect. We describe a highly-sensitive doxycycline-inducible adeno-associated virus (AAV) vector. This vector allowed for the first time a longitudinal analysis of inducible transgene expression in the brain using bioluminescence imaging. To evaluate the dose range of GDNF biological activity, the inducible AAV vector (8.0 × 10(9) viral genomes) was injected in the rat striatum at four delivery sites and increasing doxycycline doses administered orally. ERK/Akt signaling activation as well as tyrosine hydroxylase downregulation, a consequence of long-term GDNF treatment, were induced at plasmatic doxycycline concentrations of 140 and 320 ng/ml respectively, which are known not to increase antibiotic-resistant microorganisms in patients. In these conditions, GDNF covered the majority of the striatum. No behavioral abnormalities or weight loss were observed. Motor asymmetry resulting from unilateral GDNF treatment only appeared with a 2.5-fold higher vector and a 13-fold higher inducer doses. Our data suggest that using the herein-described inducible AAV vector, biological effects of GDNF can be obtained in response to sub-antimicrobial doxycycline doses.
Collapse
|
36
|
Mwangi SM, Peng S, Nezami BG, Thorn N, Farris AB, Jain S, Laroui H, Merlin D, Anania F, Srinivasan S. Glial cell line-derived neurotrophic factor protects against high-fat diet-induced hepatic steatosis by suppressing hepatic PPAR-γ expression. Am J Physiol Gastrointest Liver Physiol 2016; 310:G103-16. [PMID: 26564715 PMCID: PMC4719063 DOI: 10.1152/ajpgi.00196.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 11/02/2015] [Indexed: 01/31/2023]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) protects against high-fat diet (HFD)-induced hepatic steatosis in mice, however, the mechanisms involved are not known. In this study we investigated the effects of GDNF overexpression and nanoparticle delivery of GDNF in mice on hepatic steatosis and fibrosis and the expression of genes involved in the regulation of hepatic lipid uptake and de novo lipogenesis. Transgenic overexpression of GDNF in liver and other metabolically active tissues was protective against HFD-induced hepatic steatosis. Mice overexpressing GDNF had significantly reduced P62/sequestosome 1 protein levels suggestive of accelerated autophagic clearance. They also had significantly reduced peroxisome proliferator-activated receptor-γ (PPAR-γ) and CD36 gene expression and protein levels, and lower expression of mRNA coding for enzymes involved in de novo lipogenesis. GDNF-loaded nanoparticles were protective against short-term HFD-induced hepatic steatosis and attenuated liver fibrosis in mice with long-standing HFD-induced hepatic steatosis. They also suppressed the liver expression of steatosis-associated genes. In vitro, GDNF suppressed triglyceride accumulation in Hep G2 cells through enhanced p38 mitogen-activated protein kinase-dependent signaling and inhibition of PPAR-γ gene promoter activity. These results show that GDNF acts directly in the liver to protect against HFD-induced cellular stress and that GDNF may have a role in the treatment of nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Simon Musyoka Mwangi
- 1Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia; ,2Atlanta Veterans Affairs Medical Center, Decatur, Georgia;
| | - Sophia Peng
- 1Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia;
| | - Behtash Ghazi Nezami
- 1Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia; ,2Atlanta Veterans Affairs Medical Center, Decatur, Georgia;
| | - Natalie Thorn
- 1Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia; ,2Atlanta Veterans Affairs Medical Center, Decatur, Georgia;
| | - Alton B. Farris
- 3Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia;
| | - Sanjay Jain
- 5Internal Medicine (Renal Division), Washington University School of Medicine, St. Louis, Missouri
| | - Hamed Laroui
- 4Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia; and
| | - Didier Merlin
- 2Atlanta Veterans Affairs Medical Center, Decatur, Georgia; ,4Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia; and
| | - Frank Anania
- 1Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia; ,2Atlanta Veterans Affairs Medical Center, Decatur, Georgia;
| | - Shanthi Srinivasan
- Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia; Atlanta Veterans Affairs Medical Center, Decatur, Georgia;
| |
Collapse
|
37
|
Abstract
This chapter outlines some general principles of transcriptional targeting approaches using viral vectors in the central nervous system. Transcriptional targeting is first discussed in the context of vector tropism and appropriate delivery. Then, some of our own attempts to restrict expression of therapeutic factors to distinct brain cell populations are discussed, followed by a detailed description of the setscrews that are available for these experiments. A critical discussion of current stumbling blocks and necessary developments to achieve clinical applicability of advanced targeted vector systems is provided.
Collapse
Affiliation(s)
- Sebastian Kügler
- Department of Neurology, Center Nanoscale Microscopy and Physiology of the Brain (CNMPB), University Medicine Göttingen, Waldweg 33, 37073, Göttingen, Germany.
| |
Collapse
|
38
|
Kumar A, Kopra J, Varendi K, Porokuokka LL, Panhelainen A, Kuure S, Marshall P, Karalija N, Härma MA, Vilenius C, Lilleväli K, Tekko T, Mijatovic J, Pulkkinen N, Jakobson M, Jakobson M, Ola R, Palm E, Lindahl M, Strömberg I, Võikar V, Piepponen TP, Saarma M, Andressoo JO. GDNF Overexpression from the Native Locus Reveals its Role in the Nigrostriatal Dopaminergic System Function. PLoS Genet 2015; 11:e1005710. [PMID: 26681446 PMCID: PMC4682981 DOI: 10.1371/journal.pgen.1005710] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 11/06/2015] [Indexed: 12/14/2022] Open
Abstract
Degeneration of nigrostriatal dopaminergic system is the principal lesion in Parkinson’s disease. Because glial cell line-derived neurotrophic factor (GDNF) promotes survival of dopamine neurons in vitro and in vivo, intracranial delivery of GDNF has been attempted for Parkinson’s disease treatment but with variable success. For improving GDNF-based therapies, knowledge on physiological role of endogenous GDNF at the sites of its expression is important. However, due to limitations of existing genetic model systems, such knowledge is scarce. Here, we report that prevention of transcription of Gdnf 3’UTR in Gdnf endogenous locus yields GDNF hypermorphic mice with increased, but spatially unchanged GDNF expression, enabling analysis of postnatal GDNF function. We found that increased level of GDNF in the central nervous system increases the number of adult dopamine neurons in the substantia nigra pars compacta and the number of dopaminergic terminals in the dorsal striatum. At the functional level, GDNF levels increased striatal tissue dopamine levels and augmented striatal dopamine release and re-uptake. In a proteasome inhibitor lactacystin-induced model of Parkinson’s disease GDNF hypermorphic mice were protected from the reduction in striatal dopamine and failure of dopaminergic system function. Importantly, adverse phenotypic effects associated with spatially unregulated GDNF applications were not observed. Enhanced GDNF levels up-regulated striatal dopamine transporter activity by at least five fold resulting in enhanced susceptibility to 6-OHDA, a toxin transported into dopamine neurons by DAT. Further, we report how GDNF levels regulate kidney development and identify microRNAs miR-9, miR-96, miR-133, and miR-146a as negative regulators of GDNF expression via interaction with Gdnf 3’UTR in vitro. Our results reveal the role of GDNF in nigrostriatal dopamine system postnatal development and adult function, and highlight the importance of correct spatial expression of GDNF. Furthermore, our results suggest that 3’UTR targeting may constitute a useful tool in analyzing gene function. Intracranial delivery of GDNF has been attempted for Parkinson’s disease (PD) treatment but with variable success. For improving GDNF-based therapies, knowledge on physiological role of endogenous GDNF at the sites of its expression is important. However, due to limitations of existing genetic model systems, such knowledge is scarce. Here, we utilize an innovative genetic approach by targeting the 3’UTR regulation of Gdnf in mice. Such animals express elevated levels of Gdnf exclusively in natively Gdnf-expressing cells, enabling dissection of endogenous GDNF functions in vivo. We show that endogenous GDNF regulates dopamine system development and function and protects mice in a rodent PD model without side effects associated with ectopic GDNF applications. Further, we report how GDNF levels regulate kidney development and identify microRNAs which control GDNF expression. Our study highlights the importance of correct spatial expression of GDNF and opens a novel approach to study gene function in mice.
Collapse
Affiliation(s)
- Anmol Kumar
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Jaakko Kopra
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Kärt Varendi
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | | | - Anne Panhelainen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Satu Kuure
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Pepin Marshall
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Nina Karalija
- Department of Histology and Cell Biology, Umeå University, Umeå, Sweden
| | - Mari-Anne Härma
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Carolina Vilenius
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Kersti Lilleväli
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Triin Tekko
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Jelena Mijatovic
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Nita Pulkkinen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Madis Jakobson
- Department of Biochemistry and Developmental Biology, Institute of Biomedicine, University of Helsinki, Helsinki, Finland
| | - Maili Jakobson
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Roxana Ola
- Department of Biochemistry and Developmental Biology, Institute of Biomedicine, University of Helsinki, Helsinki, Finland
| | - Erik Palm
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Maria Lindahl
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Ingrid Strömberg
- Department of Histology and Cell Biology, Umeå University, Umeå, Sweden
| | - Vootele Võikar
- Neuroscience Center and Department of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - T. Petteri Piepponen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Jaan-Olle Andressoo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
- * E-mail:
| |
Collapse
|
39
|
Lu-Nguyen NB, Broadstock M, Yáñez-Muñoz RJ. Efficient Expression of Igf-1 from Lentiviral Vectors Protects In Vitro but Does Not Mediate Behavioral Recovery of a Parkinsonian Lesion in Rats. Hum Gene Ther 2015. [PMID: 26222254 DOI: 10.1089/hum.2015.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Gene therapy approaches delivering neurotrophic factors have offered promising results in both preclinical and clinical trials of Parkinson's disease (PD). However, failure of glial cell line-derived neurotrophic factor in phase 2 clinical trials has sparked a search for other trophic factors that may retain efficacy in the clinic. Direct protein injections of one such factor, insulin-like growth factor (IGF)-1, in a rodent model of PD has demonstrated impressive protection of dopaminergic neurons against 6-hydroxydopamine (6-OHDA) toxicity. However, protein infusion is associated with surgical risks, pump failure, and significant costs. We therefore used lentiviral vectors to deliver Igf-1, with a particular focus on the novel integration-deficient lentiviral vectors (IDLVs). A neuron-specific promoter, from the human synapsin 1 gene, excellent for gene expression from IDLVs, was additionally used to enhance Igf-1 expression. An investigation of neurotrophic effects on primary rat neuronal cultures demonstrated that neurons transduced with IDLV-Igf-1 vectors had complete protection on withdrawal of exogenous trophic support. Striatal transduction of such vectors into 6-OHDA-lesioned rats, however, provided neither protection of dopaminergic substantia nigra neurons nor improvement of animal behavior.
Collapse
Affiliation(s)
- Ngoc B Lu-Nguyen
- School of Biological Sciences, Royal Holloway, University of London , Egham, United Kingdom
| | - Martin Broadstock
- School of Biological Sciences, Royal Holloway, University of London , Egham, United Kingdom
| | - Rafael J Yáñez-Muñoz
- School of Biological Sciences, Royal Holloway, University of London , Egham, United Kingdom
| |
Collapse
|
40
|
Warren Olanow C, Bartus RT, Baumann TL, Factor S, Boulis N, Stacy M, Turner DA, Marks W, Larson P, Starr PA, Jankovic J, Simpson R, Watts R, Guthrie B, Poston K, Henderson JM, Stern M, Baltuch G, Goetz CG, Herzog C, Kordower JH, Alterman R, Lozano AM, Lang AE. Gene delivery of neurturin to putamen and substantia nigra in Parkinson disease: A double-blind, randomized, controlled trial. Ann Neurol 2015; 78:248-57. [PMID: 26061140 DOI: 10.1002/ana.24436] [Citation(s) in RCA: 183] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 04/27/2015] [Accepted: 05/04/2015] [Indexed: 11/09/2022]
Abstract
OBJECTIVE A 12-month double-blind sham-surgery-controlled trial assessing adeno-associated virus type 2 (AAV2)-neurturin injected into the putamen bilaterally failed to meet its primary endpoint, but showed positive results for the primary endpoint in the subgroup of subjects followed for 18 months and for several secondary endpoints. Analysis of postmortem tissue suggested impaired axonal transport of neurturin from putamen to substantia nigra. In the present study, we tested the safety and efficacy of AAV2-neurturin delivered to putamen and substantia nigra. METHODS We performed a 15- to 24-month, multicenter, double-blind trial in patients with advanced Parkinson disease (PD) who were randomly assigned to receive bilateral AAV2-neurturin injected bilaterally into the substantia nigra (2.0 × 10(11) vector genomes) and putamen (1.0 × 10(12) vector genomes) or sham surgery. The primary endpoint was change from baseline to final visit performed at the time the last enrolled subject completed the 15-month evaluation in the motor subscore of the Unified Parkinson's Disease Rating Scale in the practically defined off state. RESULTS Fifty-one patients were enrolled in the trial. There was no significant difference between groups in the primary endpoint (change from baseline: AAV2-neurturin, -7.0 ± 9.92; sham, -5.2 ± 10.01; p = 0.515) or in most secondary endpoints. Two subjects had cerebral hemorrhages with transient symptoms. No clinically meaningful adverse events were attributed to AAV2-neurturin. INTERPRETATION AAV2-neurturin delivery to the putamen and substantia nigra bilaterally in PD was not superior to sham surgery. The procedure was well tolerated, and there were no clinically significant adverse events related to AAV2-neurturin.
Collapse
Affiliation(s)
- C Warren Olanow
- Departments of Neurology and Neuroscience, Mount Sinai School of Medicine, New York, NY
| | | | | | - Stewart Factor
- Department of Neurology, Emory University School of Medicine, Atlanta, GA
| | - Nicholas Boulis
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA
| | - Mark Stacy
- Department of Neurology, Duke University School of Medicine, Durham, NC
| | - Dennis A Turner
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC
| | - William Marks
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Paul Larson
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Phillip A Starr
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Joseph Jankovic
- Department of Neurology, Baylor College of Medicine, Houston, TX
| | - Richard Simpson
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX
| | - Ray Watts
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL
| | - Barton Guthrie
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL
| | - Kathleen Poston
- Department of Neurology and Neurological Sciences, Stanford University Medical Center, Stanford, CA
| | - Jaimie M Henderson
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA
| | - Matthew Stern
- Department of Neurology, University of Pennsylvania, Philadelphia, PA
| | - Gordon Baltuch
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA
| | - Christopher G Goetz
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL
| | | | - Jeffrey H Kordower
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL
| | - Ron Alterman
- Beth Israel-Deaconess Medical Center, Department of Neurosurgery, Boston, MA
| | - Andres M Lozano
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Anthony E Lang
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
41
|
Viral vector delivery of neurotrophic factors for Parkinson's disease therapy. Expert Rev Mol Med 2015; 17:e8. [DOI: 10.1017/erm.2015.6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterised by the progressive loss of midbrain dopaminergic neurons, which causes motor impairments. Current treatments involve dopamine replacement to address the disease symptoms rather than its cause. Factors that promote the survival of dopaminergic neurons have been proposed as novel therapies for PD. Several dopaminergic neurotrophic factors (NTFs) have been examined for their ability to protect and/or restore degenerating dopaminergic neurons, both in animal models and in clinical trials. These include glial cell line-derived neurotrophic factor, neurturin, cerebral dopamine neurotrophic factor and growth/differentiation factor 5. Delivery of these NTFs via injection or infusion to the brain raises several practical problems. A new delivery approach for NTFs involves the use of recombinant viral vectors to enable long-term expression of these factors in brain cells. Vectors used include those based on adenoviruses, adeno-associated viruses and lentiviruses. Here we review progress to date on the potential of each of these four NTFs as novel therapeutic strategies for PD, as well as the challenges that have arisen, from pre-clinical analysis to clinical trials. We conclude by discussing recently-developed approaches to optimise the delivery of NTF-carrying viral vectors to the brain.
Collapse
|
42
|
Grabinski TM, Kneynsberg A, Manfredsson FP, Kanaan NM. A method for combining RNAscope in situ hybridization with immunohistochemistry in thick free-floating brain sections and primary neuronal cultures. PLoS One 2015; 10:e0120120. [PMID: 25794171 PMCID: PMC4368734 DOI: 10.1371/journal.pone.0120120] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 01/20/2015] [Indexed: 11/18/2022] Open
Abstract
In situ hybridization (ISH) is an extremely useful tool for localizing gene expression and changes in expression to specific cell populations in tissue samples across numerous research fields. Typically, a research group will put forth significant effort to design, generate, validate and then utilize in situ probes in thin or ultrathin paraffin embedded tissue sections. While combining ISH and IHC is an established technique, the combination of RNAscope ISH, a commercially available ISH assay with single transcript sensitivity, and IHC in thick free-floating tissue sections has not been described. Here, we provide a protocol that combines RNAscope ISH with IHC in thick free-floating tissue sections from the brain and allows simultaneous co-localization of genes and proteins in individual cells. This approach works well with a number of ISH probes (e.g. small proline-rich repeat 1a, βIII-tubulin, tau, and β-actin) and IHC antibody stains (e.g. tyrosine hydroxylase, βIII-tubulin, NeuN, and glial fibrillary acidic protein) in rat brain sections. In addition, we provide examples of combining ISH-IHC dual staining in primary neuron cultures and double-ISH labeling in thick free-floating tissue sections from the brain. Finally, we highlight the ability of RNAscope to detect ectopic DNA in neurons transduced with viral vectors. RNAscope ISH is a commercially available technology that utilizes a branched or "tree" in situ method to obtain ultrasensitive, single transcript detection. Immunohistochemistry is a tried and true method for identifying specific protein in cell populations. The combination of a sensitive and versatile oligonucleotide detection method with an established and versatile protein assay is a significant advancement in studies using free-floating tissue sections.
Collapse
Affiliation(s)
- Tessa M. Grabinski
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Andrew Kneynsberg
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Fredric P. Manfredsson
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Nicholas M. Kanaan
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
- * E-mail:
| |
Collapse
|
43
|
Wang X, Wang M, Yang L, Bai J, Yan Z, Zhang Y, Liu Z. Inhibition of Sirtuin 2 exerts neuroprotection in aging rats with increased neonatal iron intake. Neural Regen Res 2015; 9:1917-22. [PMID: 25558243 PMCID: PMC4281432 DOI: 10.4103/1673-5374.145361] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2014] [Indexed: 11/21/2022] Open
Abstract
Impaired iron homeostasis may cause damage to dopaminergic neurons and is critically involved in the pathogenesis of Parkinson's disease. At present, very little is understood about the effect of neonatal iron intake on behavior in aging animals. Therefore, we hypothesized that increased neonatal iron intake would result in significant behavior abnormalities and striatal dopamine depletion during aging, and Sirtuin 2 contributes to the age-related neurotoxicity. In the present study, we observed that neonatal iron intake (120 μg/g per day) during postnatal days 10–17 resulted in significant behavior abnormalities and striatal dopamine depletion in aging rats. Furthermore, after AK-7 (a selective Sirtuin 2 inhibitor) was injected into the substantia nigra at postnatal 540 days and 570 days (5 μg/side per day), striatal dopamine depletion was significantly diminished and behavior abnormality was improved in aging rats with neonatal iron intake. Experimental findings suggest that increased neonatal iron intake may result in Parkinson's disease-like neurochemical and behavioral deficits with aging, and inhibition of Sirtuin 2 expression may be a neuroprotective measure in Parkinson's disease.
Collapse
Affiliation(s)
- Xijin Wang
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meihua Wang
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liu Yang
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Bai
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiqiang Yan
- Shanghai Laboratory Animal Center, Chinese Academy of Sciences, Shanghai, China
| | - Yuhong Zhang
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Zhenguo Liu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
44
|
The role of parkin in the differential susceptibility of tuberoinfundibular and nigrostriatal dopamine neurons to acute toxicant exposure. Neurotoxicology 2014; 46:1-11. [PMID: 25447324 DOI: 10.1016/j.neuro.2014.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 11/07/2014] [Accepted: 11/11/2014] [Indexed: 11/22/2022]
Abstract
Parkinson disease causes degeneration of nigrostriatal dopamine (DA) neurons, while tuberoinfundibular DA neurons remain unaffected. A similar pattern is observed following exposure to 1-methy-4-phenyl-1,2,3,6-tetrahydropyradine (MPTP). The mechanism of tuberoinfundibular neuronal recovery from MPTP is associated with up-regulation of parkin protein. Here we tested if parkin mediates tuberoinfundibular neuronal recovery from MPTP by knocking-down parkin in tuberoinfundibular neurons using recombinant adeno-associated virus (rAAV), expressing a short hairpin RNA (shRNA) directed toward parkin. Following knockdown, axon terminal DA and tyrosine hydroxylase (TH) concentrations were analyzed 24h post-MPTP administration. rAAV-shRNA-mediated knockdown of endogenous parkin rendered tuberoinfundibular neurons susceptible to MPTP induced terminal DA loss, but not TH loss, within 24h post-MPTP. To determine if the neuroprotective benefits of parkin up-regulation could be translated to nigrostriatal neurons, rAAV expressing human parkin was injected into the substantia nigra of mice and axon terminal DA and TH concentrations were analyzed 24h post-MPTP. Nigral parkin over-expression prevented loss of TH in the axon terminals and soma of nigrostriatal neurons, but had no effect on terminal DA loss within 24h post-MPTP. These data show that parkin is necessary for the recovery of terminal DA concentrations within tuberoinfundibular neurons following acute MPTP administration, and parkin can rescue MPTP-induced decreases in TH within nigrostriatal neurons.
Collapse
|
45
|
Plowman EK, Thomas NJ, Kleim JA. Striatal dopamine depletion induces forelimb motor impairments and disrupts forelimb movement representations within the motor cortex. JOURNAL OF PARKINSONS DISEASE 2014; 1:93-100. [PMID: 23939260 DOI: 10.3233/jpd-2011-11017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
While limb motor deficits of Parkinson's disease are well characterized, the effects of striatal dopamine depletion on the motor cortex is poorly understood. We therefore aimed to 1) examine the effects of striatal dopamine depletion on forelimb function and cortical motor map topography and 2) explore potential relationships between forelimb function and cortical movement representations in an animal model of PD. Twenty-four male Long Evans rats were randomized to control or 6-hydroxydopamine (6-OHDA) groups. Animals in the 6-OHDA group underwent four unilateral 6-OHDA infusions into the striatum to induce striatal dopamine depletion. Four weeks later, animals were tested on a comprehensive battery of behavioral limb motor tasks followed by intracortical microstimulation to derive high-resolution topographic maps of forelimb movement representations. Standard tyrosine hydroxylase (TH) immunohistochemistry was performed and near infrared densitometry analysis utilized to assess TH depletion. Unilateral striatal dopamine depletion induced significant reductions in limb motor function that were reflected neurophysiologically as a reduction in cortical forelimb movement representations. Voluntary forelimb use, pasta handling, sunflower seed manipulation, and forelimb motor maps were all significantly impaired in 6-OHDA animals. A positive correlation was observed between forelimb function and motor map size, as well as two negative correlations between TH depletion with 1) motor map size and 2) forelimb function. The results clearly show how dysfunction within the basal ganglia thalamocortical loop resulting from nigrostriatal dopamine depletion disrupts corticospinal function.
Collapse
Affiliation(s)
- Emily K Plowman
- Department of Communication Sciences and Disorders, University of South Florida, FL 33620, U.S.A.
| | | | | |
Collapse
|
46
|
Mwangi SM, Nezami BG, Obukwelu B, Anitha M, Marri S, Fu P, Epperson MF, Le NA, Shanmugam M, Sitaraman SV, Tseng YH, Anania FA, Srinivasan S. Glial cell line-derived neurotrophic factor protects against high-fat diet-induced obesity. Am J Physiol Gastrointest Liver Physiol 2014; 306:G515-25. [PMID: 24458024 PMCID: PMC3949027 DOI: 10.1152/ajpgi.00364.2013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Obesity is a growing epidemic with limited effective treatments. The neurotrophic factor glial cell line-derived neurotrophic factor (GDNF) was recently shown to enhance β-cell mass and improve glucose control in rodents. Its role in obesity is, however, not well characterized. In this study, we investigated the ability of GDNF to protect against high-fat diet (HFD)-induced obesity. GDNF transgenic (Tg) mice that overexpress GDNF under the control of the glial fibrillary acidic protein promoter and wild-type (WT) littermates were maintained on a HFD or regular rodent diet for 11 wk, and weight gain, energy expenditure, and insulin sensitivity were monitored. Differentiated mouse brown adipocytes and 3T3-L1 white adipocytes were used to study the effects of GDNF in vitro. Tg mice resisted the HFD-induced weight gain, insulin resistance, dyslipidemia, hyperleptinemia, and hepatic steatosis seen in WT mice despite similar food intake and activity levels. They exhibited significantly (P<0.001) higher energy expenditure than WT mice and increased expression in skeletal muscle and brown adipose tissue of peroxisome proliferator activated receptor-α and β1- and β3-adrenergic receptor genes, which are associated with increased lipolysis and enhanced lipid β-oxidation. In vitro, GDNF enhanced β-adrenergic-mediated cAMP release in brown adipocytes and suppressed lipid accumulation in differentiated 3T3L-1 cells through a p38MAPK signaling pathway. Our studies demonstrate a novel role for GDNF in the regulation of high-fat diet-induced obesity through increased energy expenditure. They show that GDNF and its receptor agonists may be potential targets for the treatment or prevention of obesity.
Collapse
Affiliation(s)
- Simon Musyoka Mwangi
- 1Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia; ,2Atlanta VA Medical Center, Decatur, Georgia;
| | - Behtash Ghazi Nezami
- 1Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia;
| | - Blessing Obukwelu
- 1Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia; ,2Atlanta VA Medical Center, Decatur, Georgia;
| | - Mallappa Anitha
- 1Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia;
| | - Smitha Marri
- 1Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia;
| | - Ping Fu
- 1Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia;
| | | | - Ngoc-Anh Le
- 2Atlanta VA Medical Center, Decatur, Georgia;
| | - Malathy Shanmugam
- 3Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; and
| | - Shanthi V. Sitaraman
- 1Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia;
| | - Yu-Hua Tseng
- 4Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Frank A. Anania
- 1Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia; ,2Atlanta VA Medical Center, Decatur, Georgia;
| | - Shanthi Srinivasan
- 1Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia; ,2Atlanta VA Medical Center, Decatur, Georgia;
| |
Collapse
|
47
|
Tereshchenko J, Maddalena A, Bähr M, Kügler S. Pharmacologically controlled, discontinuous GDNF gene therapy restores motor function in a rat model of Parkinson's disease. Neurobiol Dis 2014; 65:35-42. [PMID: 24440408 DOI: 10.1016/j.nbd.2014.01.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 12/13/2013] [Accepted: 01/08/2014] [Indexed: 11/18/2022] Open
Abstract
Neurotrophic factors have raised hopes to be able to cure symptoms and to prevent progressive neurodegeneration in devastating neurological diseases. Gene therapy by means of viral vectors can overcome the hurdle of targeted delivery, but its current configuration is irreversible and thus much less controllable than that of classical pharmacotherapies. We thus aimed at developing a strategy allowing for both curative and controllable neurotrophic factor expression. Therefore, the short-term, intermittent and reversible expression of a neutrophic factor was evaluated for therapeutic efficacy in a slowly progressive animal model of Parkinson's disease (PD). We demonstrate that short-term induced expression of glial cell line derived neurotrophic factor (GDNF) is sufficient to provide i) substantial protection of nigral dopaminergic neurons from degeneration and ii) restoration of dopamine supply and motor behaviour in the partial striatal 6-OHDA model PD. These neurorestorative effects of GDNF lasted several weeks beyond the time of its expression. Later on, therapeutic efficacy ceased, but was restored by a second short induction of GDNF expression, demonstrating that monthly application of the inducing drug mifepristone was sufficient to maintain neuroprotective and neurorestorative GDNF levels. These findings suggest that forthcoming gene therapies for PD or other neurodegenerative disorders can be designed in a way that low frequency application of an approved drug can provide controllable and therapeutically efficient levels of GDNF or other neurotrophic factors. Neurotrophic factor expression can be withdrawn in case of off-target effects or sufficient clinical benefit, a feature that may eventually increase the acceptance of gene therapy for less advanced patients, which may profit better from such approaches.
Collapse
Affiliation(s)
- Julia Tereshchenko
- University Medicine Göttingen, Dept. of Neurology, Center for Molecular Physiology of the Brain, Waldweg 33, 37073 Göttingen, Germany
| | - Andrea Maddalena
- University Medicine Göttingen, Dept. of Neurology, Center for Molecular Physiology of the Brain, Waldweg 33, 37073 Göttingen, Germany
| | - Mathias Bähr
- University Medicine Göttingen, Dept. of Neurology, Center for Molecular Physiology of the Brain, Waldweg 33, 37073 Göttingen, Germany
| | - Sebastian Kügler
- University Medicine Göttingen, Dept. of Neurology, Center for Molecular Physiology of the Brain, Waldweg 33, 37073 Göttingen, Germany.
| |
Collapse
|
48
|
Douglas MR. Gene therapy for Parkinson's disease: state-of-the-art treatments for neurodegenerative disease. Expert Rev Neurother 2014; 13:695-705. [PMID: 23739006 DOI: 10.1586/ern.13.58] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Pharmacological and surgical treatments offer symptomatic benefits to patients with Parkinson's disease; however, as the condition progresses, patients experience gradual worsening in symptom control, with the development of a range of disabling complications. In addition, none of the currently available therapies have convincingly shown disease-modifying effects - either in slowing or reversing the disease. These problems have led to extensive research into the possible use of gene therapy as a treatment for Parkinson's disease. Several treatments have reached human clinical trial stages, providing important information on the risks and benefits of this novel therapeutic approach, and the tantalizing promise of improved control of this currently incurable neurodegenerative disorder.
Collapse
Affiliation(s)
- Michael R Douglas
- School of Clinical and Experimental Medicine, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
49
|
Abstract
Complex interactions between the brain and peripheral tissues mediate the effective control of energy balance and body weight. Hypothalamic and hindbrain neural circuits integrate peripheral signals informing the nutritional status of the animal and in response regulate nutrient intake and energy utilization. Obesity and its many medical complications emerge from the dysregulation of energy homeostasis. Excessive weight gain might also arise from alterations in reward systems of the brain that drive consumption of calorie dense, palatable foods in the absence of an energy requirement. Several neurotrophins, most notably brain-derived neurotrophic factor, have been implicated in the molecular and cellular processes underlying body weight regulation. Here, we review investigations interrogating their roles in energy balance and reward centers of the brain impacting feeding behavior and energy expenditure.
Collapse
Affiliation(s)
- M Rios
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, 02111, USA,
| |
Collapse
|
50
|
Chtarto A, Bockstael O, Tshibangu T, Dewitte O, Levivier M, Tenenbaum L. A next step in adeno-associated virus-mediated gene therapy for neurological diseases: regulation and targeting. Br J Clin Pharmacol 2013; 76:217-32. [PMID: 23331189 DOI: 10.1111/bcp.12065] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 12/07/2012] [Indexed: 02/04/2023] Open
Abstract
Recombinant adeno-associated virus (rAAV) vectors mediating long term transgene expression are excellent gene therapy tools for chronic neurological diseases. While rAAV2 was the first serotype tested in the clinics, more efficient vectors derived from the rh10 serotype are currently being evaluated and other serotypes are likely to be tested in the near future. In addition, aside from the currently used stereotaxy-guided intraparenchymal delivery, new techniques for global brain transduction (by intravenous or intra-cerebrospinal injections) are very promising. Various strategies for therapeutic gene delivery to the central nervous system have been explored in human clinical trials in the past decade. Canavan disease, a genetic disease caused by an enzymatic deficiency, was the first to be approved. Three gene transfer paradigms for Parkinson's disease have been explored: converting L-dopa into dopamine through AADC gene delivery in the putamen; synthesizing GABA through GAD gene delivery in the overactive subthalamic nucleus and providing neurotrophic support through neurturin gene delivery in the nigro-striatal pathway. These pioneer clinical trials demonstrated the safety and tolerability of rAAV delivery in the human brain at moderate doses. Therapeutic effects however, were modest, emphasizing the need for higher doses of the therapeutic transgene product which could be achieved using more efficient vectors or expression cassettes. This will require re-addressing pharmacological aspects, with attention to which cases require either localized and cell-type specific expression or efficient brain-wide transgene expression, and when it is necessary to modulate or terminate the administration of transgene product. The ongoing development of targeted and regulated rAAV vectors is described.
Collapse
Affiliation(s)
- Abdelwahed Chtarto
- Laboratory of Experimental Neurosurgery, Free University of Brussels (ULB), Brussels, Belgium
| | | | | | | | | | | |
Collapse
|