51
|
Time-Dependent Nerve Growth Factor Signaling Changes in the Rat Retina During Optic Nerve Crush-Induced Degeneration of Retinal Ganglion Cells. Int J Mol Sci 2017; 18:ijms18010098. [PMID: 28067793 PMCID: PMC5297732 DOI: 10.3390/ijms18010098] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 12/20/2016] [Accepted: 12/24/2016] [Indexed: 01/06/2023] Open
Abstract
Nerve growth factor (NGF) is suggested to be neuroprotective after nerve injury; however, retinal ganglion cells (RGC) degenerate following optic-nerve crush (ONC), even in the presence of increased levels of endogenous NGF. To further investigate this apparently paradoxical condition, a time-course study was performed to evaluate the effects of unilateral ONC on NGF expression and signaling in the adult retina. Visually evoked potential and immunofluorescence staining were used to assess axonal damage and RGC loss. The levels of NGF, proNGF, p75NTR, TrkA and GFAP and the activation of several intracellular pathways were analyzed at 1, 3, 7 and 14 days after crush (dac) by ELISA/Western Blot and PathScan intracellular signaling array. The progressive RGC loss and nerve impairment featured an early and sustained activation of apoptotic pathways; and GFAP and p75NTR enhancement. In contrast, ONC-induced reduction of TrkA, and increased proNGF were observed only at 7 and 14 dac. We propose that proNGF and p75NTR contribute to exacerbate retinal degeneration by further stimulating apoptosis during the second week after injury, and thus hamper the neuroprotective effect of the endogenous NGF. These findings might aid in identifying effective treatment windows for NGF-based strategies to counteract retinal and/or optic-nerve degeneration.
Collapse
|
52
|
Lu S, Han Y, Chu H, Kong L, Zhang A, Yan G, Sun H, Wang P, Wang X. Characterizing serum metabolic alterations of Alzheimer's disease and intervention of Shengmai-San by ultra-performance liquid chromatography/electrospray ionization quadruple time-of-flight mass spectrometry. Food Funct 2017; 8:1660-1671. [DOI: 10.1039/c7fo00154a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Metabolomics approach describing the nervous protective mechanism of Shengmai-San (SMS) in Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Shengwen Lu
- Heilongjiang University of Chinese Medicine
- Harbin
- China
| | - Ying Han
- Heilongjiang University of Chinese Medicine
- Harbin
- China
| | - Hang Chu
- Heilongjiang University of Chinese Medicine
- Harbin
- China
| | - Ling Kong
- Heilongjiang University of Chinese Medicine
- Harbin
- China
| | - Aihua Zhang
- Heilongjiang University of Chinese Medicine
- Harbin
- China
| | - Guangli Yan
- Heilongjiang University of Chinese Medicine
- Harbin
- China
| | - Hui Sun
- Heilongjiang University of Chinese Medicine
- Harbin
- China
| | - Ping Wang
- Heilongjiang University of Chinese Medicine
- Harbin
- China
| | - Xijun Wang
- Heilongjiang University of Chinese Medicine
- Harbin
- China
| |
Collapse
|
53
|
Janssens J, Lu D, Ni B, Chadwick W, Siddiqui S, Azmi A, Etienne H, Jushaj A, van Gastel J, Martin B, Maudsley S. Development of Precision Small-Molecule Proneurotrophic Therapies for Neurodegenerative Diseases. VITAMINS AND HORMONES 2016; 104:263-311. [PMID: 28215298 DOI: 10.1016/bs.vh.2016.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Age-related neurodegenerative diseases, such as Alzheimer's disease, will represent one of the largest future burdens on worldwide healthcare systems due to the increasing proportion of elderly in our society. As deficiencies in neurotrophins are implicated in the pathogenesis of many age-related neurodegenerative disorders, it is reasonable to consider that global neurotrophin resistance may also become a major healthcare threat. Central nervous system networks are effectively maintained through aging by neuroprotective and neuroplasticity signaling mechanisms which are predominantly controlled by neurotrophin receptor signaling. Neurotrophin receptors are single pass receptor tyrosine kinases that form dimeric structures upon ligand binding to initiate cellular signaling events that control many protective and plasticity-related pathways. Declining functionality of the neurotrophin ligand-receptor system is considered one of the hallmarks of neuropathological aging. Therefore, it is imperative to develop effective therapeutic strategies to contend with this significant issue. While the therapeutic applications of cognate ligands for neurotrophin receptors are limited, the development of nonpeptidergic, small-molecule ligands can overcome these limitations, and productively regulate this important receptor system with beneficial effects. Using our advanced knowledge of the high-dimensionality complexity of receptor systems, the future generation of precision medicines targeting these systems will be an attainable goal.
Collapse
Affiliation(s)
- J Janssens
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - D Lu
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - B Ni
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - W Chadwick
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - S Siddiqui
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - A Azmi
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - H Etienne
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - A Jushaj
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - J van Gastel
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - B Martin
- Metabolism Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - S Maudsley
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium; Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States.
| |
Collapse
|
54
|
Hu R, Jin S, He X, Xu F, Hu J. Whole-Brain Monosynaptic Afferent Inputs to Basal Forebrain Cholinergic System. Front Neuroanat 2016; 10:98. [PMID: 27777554 PMCID: PMC5056182 DOI: 10.3389/fnana.2016.00098] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 09/28/2016] [Indexed: 12/17/2022] Open
Abstract
The basal forebrain cholinergic system (BFCS) robustly modulates many important behaviors, such as arousal, attention, learning and memory, through heavy projections to cortex and hippocampus. However, the presynaptic partners governing BFCS activity still remain poorly understood. Here, we utilized a recently developed rabies virus-based cell-type-specific retrograde tracing system to map the whole-brain afferent inputs of the BFCS. We found that the BFCS receives inputs from multiple cortical areas, such as orbital frontal cortex, motor cortex, and insular cortex, and that the BFCS also receives dense inputs from several subcortical nuclei related to motivation and stress, including lateral septum, central amygdala, paraventricular nucleus of hypothalamus, dorsal raphe, and parabrachial nucleus. Interestingly, we found that the BFCS receives inputs from the olfactory areas and the entorhinal–hippocampal system. These results greatly expand our knowledge about the connectivity of the mouse BFCS and provided important preliminary indications for future exploration of circuit function.
Collapse
Affiliation(s)
- Rongfeng Hu
- Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University Xi'an, China
| | - Sen Jin
- Center for Excellence in Brain Science, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences Wuhan, China
| | - Xiaobin He
- Center for Excellence in Brain Science, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences Wuhan, China
| | - Fuqiang Xu
- Center for Excellence in Brain Science, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences Wuhan, China
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University Shanghai, China
| |
Collapse
|
55
|
Josephy-Hernandez S, Jmaeff S, Pirvulescu I, Aboulkassim T, Saragovi HU. Neurotrophin receptor agonists and antagonists as therapeutic agents: An evolving paradigm. Neurobiol Dis 2016; 97:139-155. [PMID: 27546056 DOI: 10.1016/j.nbd.2016.08.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 08/10/2016] [Accepted: 08/16/2016] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative disorders are prevalent, complex and devastating conditions, with very limited treatment options currently available. While they manifest in many forms, there are commonalities that link them together. In this review, we will focus on neurotrophins - a family of related factors involved in neuronal development and maintenance. Neurodegenerative diseases often present with a neurotrophin imbalance, in which there may be decreases in trophic signaling through Trk receptors for example, and/or increases in pro-apoptotic activity through p75. Clinical trials with neurotrophins have continuously failed due to their poor pharmacological properties as well as the unavoidable activation of p75. Thus, there is a need for drugs without such setbacks. Small molecule neurotrophin mimetics are favorable options since they can selectively activate Trks or inactivate p75. In this review, we will initially present a brief outline of how these molecules are synthesized and their mechanisms of action; followed by an update in the current state of neurotrophins and small molecules in major neurodegenerative diseases. Although there has been significant progress in the development of potential therapeutics, more studies are needed to establish clear mechanisms of action and target specificity in order to transition from animal models to the assessment of safety and use in humans.
Collapse
Affiliation(s)
- Sylvia Josephy-Hernandez
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Sean Jmaeff
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Iulia Pirvulescu
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Tahar Aboulkassim
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - H Uri Saragovi
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
56
|
Eyjolfsdottir H, Eriksdotter M, Linderoth B, Lind G, Juliusson B, Kusk P, Almkvist O, Andreasen N, Blennow K, Ferreira D, Westman E, Nennesmo I, Karami A, Darreh-Shori T, Kadir A, Nordberg A, Sundström E, Wahlund LO, Wall A, Wiberg M, Winblad B, Seiger Å, Wahlberg L, Almqvist P. Targeted delivery of nerve growth factor to the cholinergic basal forebrain of Alzheimer's disease patients: application of a second-generation encapsulated cell biodelivery device. ALZHEIMERS RESEARCH & THERAPY 2016; 8:30. [PMID: 27389402 PMCID: PMC4936020 DOI: 10.1186/s13195-016-0195-9] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 06/06/2016] [Indexed: 12/21/2022]
Abstract
Background Targeted delivery of nerve growth factor (NGF) has emerged as a potential therapy for Alzheimer’s disease (AD) due to its regenerative effects on basal forebrain cholinergic neurons. This hypothesis has been tested in patients with AD using encapsulated cell biodelivery of NGF (NGF-ECB) in a first-in-human study. We report our results from a third-dose cohort of patients receiving second-generation NGF-ECB implants with improved NGF secretion. Methods Four patients with mild to moderate AD were recruited to participate in an open-label, phase Ib dose escalation study with a 6-month duration. Each patient underwent stereotactic implant surgery with four NGF-ECB implants targeted at the cholinergic basal forebrain. The NGF secretion of the second-generation implants was improved by using the Sleeping Beauty transposon gene expression technology and an improved three-dimensional internal scaffolding, resulting in production of about 10 ng NGF/device/day. Results All patients underwent successful implant procedures without complications, and all patients completed the study, including implant removal after 6 months. Upon removal, 13 of 16 implants released NGF, 8 implants released NGF at the same rate or higher than before the implant procedure, and 3 implants failed to release detectable amounts of NGF. Of 16 adverse events, none was NGF-, or implant-related. Changes from baseline values of cholinergic markers in cerebrospinal fluid (CSF) correlated with cortical nicotinic receptor expression and Mini Mental State Examination score. Levels of neurofilament light chain (NFL) protein increased in CSF after NGF-ECB implant, while glial fibrillary acidic protein (GFAP) remained stable. Conclusions The data derived from this patient cohort demonstrate the safety and tolerability of sustained NGF release by a second-generation NGF-ECB implant to the basal forebrain, with uneventful surgical implant and removal of NGF-ECB implants in a new dosing cohort of four patients with AD. Trial registration ClinicalTrials.gov identifier: NCT01163825. Registered on 14 Jul 2010.
Collapse
Affiliation(s)
- Helga Eyjolfsdottir
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden.,Department of Geriatrics, Karolinska University Hospital, Huddinge, 171 76, Stockholm, Sweden
| | - Maria Eriksdotter
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden.,Department of Geriatrics, Karolinska University Hospital, Huddinge, 171 76, Stockholm, Sweden
| | - Bengt Linderoth
- Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden.,Department of Neurosurgery, Karolinska University Hospital Solna, Building R3:02, 171 76, Stockholm, Sweden
| | - Göran Lind
- Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden.,Department of Neurosurgery, Karolinska University Hospital Solna, Building R3:02, 171 76, Stockholm, Sweden
| | - Bengt Juliusson
- NsGene Inc., 225 Chapman Street, Providence, RI, 02905-4533, USA
| | - Philip Kusk
- NsGene Inc., 225 Chapman Street, Providence, RI, 02905-4533, USA
| | - Ove Almkvist
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Niels Andreasen
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden.,Department of Geriatrics, Karolinska University Hospital, Huddinge, 171 76, Stockholm, Sweden
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Department of Clinical Neuroscience, University of Gothenburg, 41345, Gothenburg, Sweden
| | - Daniel Ferreira
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Eric Westman
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Inger Nennesmo
- Department of Laboratory Medicine, Section of Pathology, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Azadeh Karami
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Taher Darreh-Shori
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Ahmadul Kadir
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Agneta Nordberg
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden.,Department of Geriatrics, Karolinska University Hospital, Huddinge, 171 76, Stockholm, Sweden
| | - Erik Sundström
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden.,Stiftelsen Stockholms Sjukhem, Mariebergsgatan 22, 112 35, Stockholm, Sweden
| | - Lars-Olof Wahlund
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden.,Department of Geriatrics, Karolinska University Hospital, Huddinge, 171 76, Stockholm, Sweden
| | - Anders Wall
- Department of Surgical Sciences, Section of Nuclear Medicine and PET, Uppsala University Hospital, 75185, Uppsala, Sweden
| | - Maria Wiberg
- Department of Clinical Science, Intervention and Technology, Division of Medical Imaging and Technology, Karolinska Institutet, 171 77, Stockholm, Sweden.,Department of Radiology, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden.,Department of Geriatrics, Karolinska University Hospital, Huddinge, 171 76, Stockholm, Sweden
| | - Åke Seiger
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden.,Stiftelsen Stockholms Sjukhem, Mariebergsgatan 22, 112 35, Stockholm, Sweden
| | - Lars Wahlberg
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden.,NsGene Inc., 225 Chapman Street, Providence, RI, 02905-4533, USA
| | - Per Almqvist
- Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden. .,Department of Neurosurgery, Karolinska University Hospital Solna, Building R3:02, 171 76, Stockholm, Sweden.
| |
Collapse
|
57
|
The Emerging Therapeutic Role of NGF in Alzheimer’s Disease. Neurochem Res 2016; 41:1211-8. [DOI: 10.1007/s11064-016-1829-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/08/2015] [Accepted: 01/05/2016] [Indexed: 11/29/2022]
|
58
|
Soligo M, Protto V, Florenzano F, Bracci-Laudiero L, De Benedetti F, Chiaretti A, Manni L. The mature/pro nerve growth factor ratio is decreased in the brain of diabetic rats: Analysis by ELISA methods. Brain Res 2015; 1624:455-468. [PMID: 26282349 DOI: 10.1016/j.brainres.2015.08.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 07/21/2015] [Accepted: 08/08/2015] [Indexed: 01/08/2023]
Abstract
Nerve growth factor (NGF) is essential for the survival and functional maintenance of forebrain cholinergic neurons projecting mainly to the cortex and hippocampus. NGF is produced in these brain areas but while mature NGF (mNGF) has a survival/differentiative effect its precursor proNGF elicits apoptosis in cholinergic neurons. Impaired neurotransmission, loss of cholinergic phenotype and abnormal NGF content characterize the cholinergic circuitries in animal models of diabetic encephalopathy (DE). It is not known whether defective production or maturation of NGF could play a key role in cholinergic neurodegeneration in DE. Quantification of the mNGF/proNGF ratio is therefore needed to characterize the development and progression of NGF-related neuronal diseases. In our work, we aimed at developing ELISA methods to measure either mNGF or proNGF tissue concentration; and to define the mNGF/proNGF ratio in the rat cortex and hippocampus during the early stage of streptozotocin-induced type 1 diabetes. Using commercially available NGF ELISA kits and antibodies, we set up ELISAs for human and rat mNGF and proNGF. We then analyzed the mNGF/proNGF ratio in the cortex and hippocampus of DE rats and found that it decreased in both tissues starting from the fourth week after diabetes induction. In diabetic brain the increase in proNGF involves accumulation of the isoforms with molecular weights of 50 and 34 kDa. Our study for the first time specifically quantifies the absolute content of mature and proNGF and the mNGF/proNGF ratio in brain tissues, suggesting that early progression of experimental DE is characterized by defective maturation of NGF.
Collapse
Affiliation(s)
- Marzia Soligo
- Institute of Translational Pharmacology-CNR, via del Fosso del Cavaliere 100, 00133 Rome, Italy
| | - Virginia Protto
- Institute of Translational Pharmacology-CNR, via del Fosso del Cavaliere 100, 00133 Rome, Italy
| | - Fulvio Florenzano
- Confocal Microscopy Unit, European Brain Research Institute (EBRI), Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - Luisa Bracci-Laudiero
- Institute of Translational Pharmacology-CNR, via del Fosso del Cavaliere 100, 00133 Rome, Italy; Bambino Gesu' Children's Hospital-IRCSS, Division of Rheumatology, Piazza S. Onofrio 4, 00165 Rome, Italy
| | - Fabrizio De Benedetti
- Bambino Gesu' Children's Hospital-IRCSS, Division of Rheumatology, Piazza S. Onofrio 4, 00165 Rome, Italy
| | - Antonio Chiaretti
- Department of Pediatrics, Catholic University of Rome, Largo A Gemelli 8, 00168 Rome, Italy
| | - Luigi Manni
- Institute of Translational Pharmacology-CNR, via del Fosso del Cavaliere 100, 00133 Rome, Italy.
| |
Collapse
|
59
|
Tirassa P, Maccarone M, Carito V, De Nicolò S, Fiore M. Ocular nerve growth factor administration counteracts the impairment of neural precursor cell viability and differentiation in the brain subventricular area of rats with streptozotocin-induced diabetes. Eur J Neurosci 2015; 41:1207-18. [PMID: 25728260 DOI: 10.1111/ejn.12854] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 01/07/2015] [Accepted: 01/19/2015] [Indexed: 01/12/2023]
Abstract
The ocular administration of nerve growth factor (NGF) as eye drops (oNGF) has been shown to exert protective effects in forebrain-injured animal models, including adult diabetes induced by a single injection of streptozotocin (STZ) (60 mg/kg body weight). This type 1 diabetes model was used in this study to investigate whether oNGF might extend its actions on neuronal precursors localised in the subventricular zone (SVZ). NGF or saline was administrated as eye drops twice daily for 2 weeks in rats with STZ-induced diabetes and healthy control rats. The expression of mature and precursor NGF and the NGF receptors, tropomyosin-related kinase A and neurotrophin receptor p75, and the levels of DNA fragmentation were analysed by ELISA and western blotting. Incorporation of bromodeoxyuridine was used to trace newly formed cells. Nestin, polysialylated neuronal cell adhesion molecule (PSA-NCAM), doublecortin (DCX) and glial fibrillary acidic protein antibodies were used to identify the SVZ cells by confocal microscopy. It was found that oNGF counteracts the STZ-induced cell death and the alteration of mature/pro-NGF expression in the SVZ. It also affects the survival and differentiation of SVZ progenitors. In particular, oNGF counteracts the reduction in the number of cells expressing PSA-NCAM/DCX (neuroblast type A cells) and the related reductions in the number and distribution of nestin/DCX-positive cells (C-type cells), or glia-committed cells (type B cells), observed in the SVZ of diabetic rats. These findings show that oNGF treatment counteracts the effect of type 1 diabetes on neuronal precursors in the SVZ, and further support the neuroprotective and reparative role of oNGF in the brain.
Collapse
Affiliation(s)
- Paola Tirassa
- Institute of Cell Biology and Neurobiology, National Research Council (CNR), Via del Fosso di Fiorano, 64 (00143), Rome, Italy
| | | | | | | | | |
Collapse
|
60
|
Karami A, Eyjolfsdottir H, Vijayaraghavan S, Lind G, Almqvist P, Kadir A, Linderoth B, Andreasen N, Blennow K, Wall A, Westman E, Ferreira D, Kristoffersen Wiberg M, Wahlund LO, Seiger Å, Nordberg A, Wahlberg L, Darreh-Shori T, Eriksdotter M. Changes in CSF cholinergic biomarkers in response to cell therapy with NGF in patients with Alzheimer's disease. Alzheimers Dement 2015; 11:1316-28. [PMID: 25676388 DOI: 10.1016/j.jalz.2014.11.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 09/26/2014] [Accepted: 11/20/2014] [Indexed: 12/31/2022]
Abstract
INTRODUCTION The extensive loss of central cholinergic functions in Alzheimer's disease (AD) brain is linked to impaired nerve growth factor (NGF) signaling. The cardinal cholinergic biomarker is the acetylcholine synthesizing enzyme, choline acetyltransferase (ChAT), which has recently been found in cerebrospinal fluid (CSF). The purpose of this study was to see if EC-NGF therapy will alter CSF levels of cholinergic biomarkers, ChAT, and acetylcholinesterase. METHOD Encapsulated cell implants releasing NGF (EC-NGF) were surgically implanted bilaterally in the basal forebrain of six AD patients for 12 months and cholinergic markers in CSF were analyzed. RESULTS Activities of both enzymes were altered after 12 months. In particular, the activity of soluble ChAT showed high correlation with cognition, CSF tau and amyloid-β, in vivo cerebral glucose utilization and nicotinic binding sites, and morphometric and volumetric magnetic resonance imaging measures. DISCUSSION A clear pattern of association is demonstrated showing a proof-of-principle effect on CSF cholinergic markers, suggestive of a beneficial EC-NGF implant therapy.
Collapse
Affiliation(s)
- Azadeh Karami
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Geriatrics, Karolinska University Hospital, Stockholm, Sweden
| | - Helga Eyjolfsdottir
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Geriatrics, Karolinska University Hospital, Stockholm, Sweden
| | - Swetha Vijayaraghavan
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Göran Lind
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden
| | - Per Almqvist
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden
| | - Ahmadul Kadir
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Linderoth
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden
| | - Niels Andreasen
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Geriatrics, Karolinska University Hospital, Stockholm, Sweden
| | - Kaj Blennow
- Department of Clinical Neuroscience, Clinical Neurochemistry Laboratory, University of Göteborg, Göteborg, Sweden
| | - Anders Wall
- Nuclear medicine and PET, Department of Surgical Sciences, Uppsala University, Sweden
| | - Eric Westman
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Geriatrics, Karolinska University Hospital, Stockholm, Sweden
| | - Daniel Ferreira
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Maria Kristoffersen Wiberg
- Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden; Department of Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Lars-Olof Wahlund
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Geriatrics, Karolinska University Hospital, Stockholm, Sweden
| | | | - Agneta Nordberg
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Geriatrics, Karolinska University Hospital, Stockholm, Sweden
| | | | - Taher Darreh-Shori
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Geriatrics, Karolinska University Hospital, Stockholm, Sweden.
| | - Maria Eriksdotter
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Geriatrics, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
61
|
Simmons DA, Knowles JK, Belichenko NP, Banerjee G, Finkle C, Massa SM, Longo FM. A small molecule p75NTR ligand, LM11A-31, reverses cholinergic neurite dystrophy in Alzheimer's disease mouse models with mid- to late-stage disease progression. PLoS One 2014; 9:e102136. [PMID: 25153701 PMCID: PMC4143160 DOI: 10.1371/journal.pone.0102136] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 06/16/2014] [Indexed: 11/18/2022] Open
Abstract
Degeneration of basal forebrain cholinergic neurons contributes significantly to the cognitive deficits associated with Alzheimer's disease (AD) and has been attributed to aberrant signaling through the neurotrophin receptor p75 (p75NTR). Thus, modulating p75NTR signaling is considered a promising therapeutic strategy for AD. Accordingly, our laboratory has developed small molecule p75NTR ligands that increase survival signaling and inhibit amyloid-β-induced degenerative signaling in in vitro studies. Previous work found that a lead p75NTR ligand, LM11A-31, prevents degeneration of cholinergic neurites when given to an AD mouse model in the early stages of disease pathology. To extend its potential clinical applications, we sought to determine whether LM11A-31 could reverse cholinergic neurite atrophy when treatment begins in AD mouse models having mid- to late stages of pathology. Reversing pathology may have particular clinical relevance as most AD studies involve patients that are at an advanced pathological stage. In this study, LM11A-31 (50 or 75 mg/kg) was administered orally to two AD mouse models, Thy-1 hAPPLond/Swe (APPL/S) and Tg2576, at age ranges during which marked AD-like pathology manifests. In mid-stage male APPL/S mice, LM11A-31 administered for 3 months starting at 6-8 months of age prevented and/or reversed atrophy of basal forebrain cholinergic neurites and cortical dystrophic neurites. Importantly, a 1 month LM11A-31 treatment given to male APPL/S mice (12-13 months old) with late-stage pathology reversed the degeneration of cholinergic neurites in basal forebrain, ameliorated cortical dystrophic neurites, and normalized increased basal forebrain levels of p75NTR. Similar results were seen in female Tg2576 mice. These findings suggest that LM11A-31 can reduce and/or reverse fundamental AD pathologies in late-stage AD mice. Thus, targeting p75NTR is a promising approach to reducing AD-related degenerative processes that have progressed beyond early stages.
Collapse
Affiliation(s)
- Danielle A. Simmons
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, United States of America
| | - Juliet K. Knowles
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, United States of America
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Nadia P. Belichenko
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, United States of America
| | - Gargi Banerjee
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, United States of America
| | - Carly Finkle
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, United States of America
| | - Stephen M. Massa
- Department of Neurology and Laboratory for Computational Neurochemistry and Drug Discovery, San Francisco Veterans Affairs Medical Center, and Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Frank M. Longo
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, United States of America
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
62
|
Mele T, Jurič DM. Metrifonate, like acetylcholine, up-regulates neurotrophic activity of cultured rat astrocytes. Pharmacol Rep 2014; 66:618-23. [DOI: 10.1016/j.pharep.2014.02.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 02/03/2014] [Accepted: 02/26/2014] [Indexed: 12/26/2022]
|
63
|
Angelucci F, Gelfo F, Fiore M, Croce N, Mathé AA, Bernardini S, Caltagirone C. The effect of neuropeptide Y on cell survival and neurotrophin expression in in-vitro models of Alzheimer's disease. Can J Physiol Pharmacol 2014; 92:621-30. [PMID: 25026432 DOI: 10.1139/cjpp-2014-0099] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a disorder characterized by the accumulation of abnormally folded protein fragments in neurons, i.e., β-amyloid (Aβ) and tau protein, leading to cell death. Several neuropeptides present in the central nervous system (CNS) are believed to be involved in the pathophysiology of AD. Among them, neuropeptide Y (NPY), a small peptide widely distributed throughout the brain, has generated interest because of its role in neuroprotection against excitotoxicity in animal models of AD. In addition, it has been shown that NPY modulates neurogenesis. Interestingly, these latter effects are similar to those elicited by neurotrophins, which are critical molecules for the function and survival of neurons that degenerate during the course of AD. In this review we summarize the evidence for the involvement of NPY and neurotrophins in AD pathogenesis, and the similarity between them in CNS neurons. Finally, we recapitulate our recent in-vitro evidence for the involvement of neurotrophin nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) in the neuroprotective effect elicited by NPY in AD neuron-like models (neuroblastoma cells or primary cultures exposed to toxic concentrations of Aβ's pathogenic fragment 25-35), and propose a putative mechanism based on NPY-induced inhibition of voltage-dependent Ca(2+) influx in pre- and post-synaptic neurons.
Collapse
Affiliation(s)
- Francesco Angelucci
- a Department of Clinical and Behavioural Neurology, IRCCS Santa Lucia Foundation, Via Ardeatina 354, 00142 Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
64
|
Kniewallner KM, Grimm N, Humpel C. Platelet-derived nerve growth factor supports the survival of cholinergic neurons in organotypic rat brain slices. Neurosci Lett 2014; 574:64-9. [PMID: 24861506 DOI: 10.1016/j.neulet.2014.05.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/07/2014] [Accepted: 05/14/2014] [Indexed: 12/12/2022]
Abstract
Platelets play a role in repair of vessels and contain different growth factors, including nerve growth factor (NGF). Since NGF is the most potent growth factor to support survival of cholinergic neurons, we aimed to study the effects of platelet-derived NGF on cholinergic neurons in organotypic brain slices. Brain slices of the nucleus basalis of Meynert (nBM) were cultured with or without NGF (10ng/ml) or platelet extracts (100μg/ml) or fresh platelets (10(8) platelets/ml). In order to enhance NGF in platelets recombinant NGF (100ng) was loaded into platelets using ultrasound (3h). Our data show that recombinant NGF markedly supports survival of cholinergic neurons. The addition of fresh platelets showed a tendency for enhancing cholinergic neuron numbers, while platelet extracts had no effects. Ultrasound was highly effective to load recombinant NGF into platelets. The addition of NGF-loaded platelets markedly enhanced cholinergic neuron numbers. In conclusion, our data provide evidence that NGF-derived platelets may counteract cell death of cholinergic neurons.
Collapse
Affiliation(s)
- Kathrin M Kniewallner
- Laboratory of Psychiatry and Exp. Alzheimer Resaerch, Department of Psychiatry and Psychotherapy, Innsbruck Medical University, Anichstr. 35, A-6020 Innsbruck, Austria
| | - Natalia Grimm
- Laboratory of Psychiatry and Exp. Alzheimer Resaerch, Department of Psychiatry and Psychotherapy, Innsbruck Medical University, Anichstr. 35, A-6020 Innsbruck, Austria
| | - Christian Humpel
- Laboratory of Psychiatry and Exp. Alzheimer Resaerch, Department of Psychiatry and Psychotherapy, Innsbruck Medical University, Anichstr. 35, A-6020 Innsbruck, Austria.
| |
Collapse
|
65
|
Non-noxious skin stimulation activates the nucleus basalis of Meynert and promotes NGF secretion in the parietal cortex via nicotinic ACh receptors. J Physiol Sci 2014; 64:253-60. [PMID: 24801530 PMCID: PMC4070488 DOI: 10.1007/s12576-014-0313-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 03/20/2014] [Indexed: 02/01/2023]
Abstract
The effects of non-noxious skin stimulation on nerve growth factor (NGF) secretion in the parietal cortex were examined in anesthetized rats. Innocuous skin stimulation was delivered to the left hindlimb with a soft-hair brush. Extracellular NGF in the right parietal cortex was collected by microdialysis methods using a protein-permeable probe and was measured using an enzyme-linked immune-sorbent assay. Brushing produced a significant increase in extracellular NGF levels. This NGF response was not observed in rats pretreated with a nicotinic ACh receptor (nAChR) antagonist mecamylamine. We further examined whether brushing could activate the basal forebrain nucleus (nucleus basalis of Meynert, NBM), which is the main source of cholinergic fibers in the cerebral cortex, by means of functional MRI. The blood oxygen level-dependent signal in the right NBM was significantly higher during brushing compared to baseline. The results suggest that non-noxious skin stimulation activates NBM and promotes NGF secretion in the parietal cortex via nAChRs.
Collapse
|
66
|
Arisi I, D'Onofrio M, Brandi R, Malerba F, Paoletti F, Storti AE, Florenzano F, Fasulo L, Cattaneo A. proNGF/NGF mixtures induce gene expression changes in PC12 cells that neither singly produces. BMC Neurosci 2014; 15:48. [PMID: 24713110 PMCID: PMC4098786 DOI: 10.1186/1471-2202-15-48] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 03/27/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Growing evidence shows that, in vivo, the precursor of Nerve Growth Factor (NGF), proNGF, displays biological activities different from those of its mature NGF counterpart, mediated by distinct, and somewhat complementary, receptor binding properties. NGF and proNGF induce distinct transcriptional signatures in target cells, highlighting their different bioactivities. In vivo, proNGF and mature NGF coexist. It was proposed that the relative proNGF/NGF ratio is important for their biological outcomes, especially in pathological conditions, since proNGF, the principal form of NGF in Central Nervous System (CNS), is increased in Alzheimer's disease brains. These observations raise a relevant question: does proNGF, in the presence of NGF, influence the NGF transcriptional response and viceversa? In order to understand the specific proNGF effect on NGF activity, depending on the relative proNGF/NGF concentration, we investigated whether proNGF affects the pattern of well-known NGF-regulated mRNAs. RESULTS To test any influence of proNGF on pure NGF expression fingerprinting, the expression level of a set of candidate genes was analysed by qReal-Time PCR in rat adrenal pheochromocytoma cell line PC12, treated with a mixture of NGF and proNGF recombinant proteins, in different stoichiometric ratios. These candidates were selected amongst a set of genes well-known as being rapidly induced by NGF treatment. We found that, when PC12 cells are treated with proNGF/NGF mixtures, a unique pattern of gene expression, which does not overlap with that deriving from treatment with either proNGF or NGF alone, is induced. The specific effect is also dependent on the stoichiometric composition of the mixture. The proNGF/NGF equimolar mixture seems to partially neutralize the specific effects of the proNGF or NGF individual treatments, showing a weaker overall response, compared to the individual contributions of NGF and proNGF alone. CONCLUSIONS Using gene expression as a functional read-out, our data demonstrate that the relative availability of NGF and proNGF in vivo might modulate the biological outcome of these ligands.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Antonino Cattaneo
- Neurotrophic Factors and Neurodegenerative Diseases Laboratory, European Brain Research Institute (EBRI) "Rita Levi-Montalcini", Via del Fosso di Fiorano, 64, 00143 Roma, Italy.
| |
Collapse
|
67
|
Fields J, Dumaop W, Langford TD, Rockenstein E, Masliah E. Role of neurotrophic factor alterations in the neurodegenerative process in HIV associated neurocognitive disorders. J Neuroimmune Pharmacol 2014; 9:102-16. [PMID: 24510686 PMCID: PMC3973421 DOI: 10.1007/s11481-013-9520-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 12/26/2013] [Indexed: 12/30/2022]
Abstract
Migration of HIV infected cells into the CNS is associated with a spectrum of neurological disorders, ranging from milder forms of HIV-associated neurocognitive disorders (HAND) to HIV-associated dementia (HAD). These neuro-psychiatric syndromes are related to the neurodegenerative pathology triggered by the release of HIV proteins and cytokine/chemokines from monocytes/macrophages into the CNS -a condition known as HIV encephalitis (HIVE). As a result of more effective combined anti-retroviral therapy patients with HIV are living longer and thus the frequency of HAND has increased considerably, resulting in an overlap between the neurodegenerative pathology associated with HIV and that related to aging. In fact, HIV infection is believed to hasten the aging process. The mechanisms through which HIV and aging lead to neurodegeneration include: abnormal calcium flux, excitotoxicity, signaling abnormalities, oxidative stress and autophagy defects. Moreover, recent studies have shown that defects in the processing and transport of neurotrophic factors such as fibroblast growth factors (FGFs), neural growth factor (NGF) and brain-derived growth factor (BDNF) might also play a role. Recent evidence implicates alterations in neurotrophins in the pathogenesis of neurodegeneration associated with HAND in the context of aging. Here, we report FGF overexpression curtails gp120-induced neurotoxicity in a double transgenic mouse model. Furthermore, our data show disparities in brain neurotrophic factor levels may be exacerbated in HIV patients over 50 years of age. In this review, we discuss the most recent findings on neurotrophins and HAND in the context of developing new therapies to combat HIV infection in the aging population.
Collapse
Affiliation(s)
- Jerel Fields
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | | | | | | | | |
Collapse
|
68
|
Transient disturbances in contextual fear memory induced by Aβ(25–35) in rats are accompanied by cholinergic dysfunction. Behav Brain Res 2014; 259:152-7. [DOI: 10.1016/j.bbr.2013.11.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 11/07/2013] [Accepted: 11/10/2013] [Indexed: 11/22/2022]
|
69
|
Wang W, Chen J, Guo X. The role of nerve growth factor and its receptors in tumorigenesis and cancer pain. Biosci Trends 2014; 8:68-74. [DOI: 10.5582/bst.8.68] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
70
|
Deep brain stimulation in dementia-related disorders. Neurosci Biobehav Rev 2013; 37:2666-75. [DOI: 10.1016/j.neubiorev.2013.09.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 08/21/2013] [Accepted: 09/03/2013] [Indexed: 11/21/2022]
|
71
|
Duan L, Chen BY, Sun XL, Luo ZJ, Rao ZR, Wang JJ, Chen LW. LPS-induced proNGF synthesis and release in the N9 and BV2 microglial cells: a new pathway underling microglial toxicity in neuroinflammation. PLoS One 2013; 8:e73768. [PMID: 24040063 PMCID: PMC3767823 DOI: 10.1371/journal.pone.0073768] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 07/23/2013] [Indexed: 01/22/2023] Open
Abstract
Purpose While aberrant activation of microglial cells was evidently involved in neuroinflammation and neurotoxicity in the neurodegenerative diseases such as Alzheimer’s and Parkinson’s disease, objective of study was to address if activated microglias deliver their effect by releasing pro-neurotrophins. Materials and methods By in vitro culture of N9 and BV2 cell lines and lipopolysaccharide (LPS) stimulation model, generation and release of proNGF, proBDNF and MMP-9 was studied in the activated microglial cells by immunocytochemistry, western blotting and bioassay methods. Results Activation of microglial cells was observed with obvious increasing iba1-immunoreactivity following LPS stimulation in cell culture. Synthesis and up-regulation of proNGF protein significantly occurred in N9 and BV2 cells 12h-48h after LPS exposure, whereas no significant changes of proBDNF and MMP9 were observed in these microglial cell lines with LPS insult. More interestingly, extracellular release or secretion of proNGF molecule was also detected in culture medium of N9 cells after LPS stimulation. Finally, bioassay using MTT, Hoechst/PI and TUNEL staining in SH-SY5Y cells further confirmed that proNGF treatment could result in apoptotic cell death but it did not significantly influence cell viability of SH-SY5Y cells. Conclusions This in vitro study revealed LPS-stimulated proNGF synthesis and release in activated N9/BV2 microglial cell lines, also suggesting that proNGF may appeal a new pathway or possible mechanism underlying microglial toxicity in the neuroinflammation and a potential target for therapeutic manipulation of the neurodegenerative diseases.
Collapse
Affiliation(s)
- Li Duan
- Institute of Neurosciences, The Fourth Military Medical University, Xi’an, China
| | - Bei-Yu Chen
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Xiao-Long Sun
- Institute of Neurosciences, The Fourth Military Medical University, Xi’an, China
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Zhuo-Jing Luo
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Zhi-Ren Rao
- Institute of Neurosciences, The Fourth Military Medical University, Xi’an, China
| | - Jing-Jie Wang
- Department of Gastroenterology, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
- * E-mail: (JW); (LC)
| | - Liang-Wei Chen
- Institute of Neurosciences, The Fourth Military Medical University, Xi’an, China
- * E-mail: (JW); (LC)
| |
Collapse
|
72
|
Small-molecule modulation of neurotrophin receptors: a strategy for the treatment of neurological disease. Nat Rev Drug Discov 2013; 12:507-25. [PMID: 23977697 DOI: 10.1038/nrd4024] [Citation(s) in RCA: 198] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Neurotrophins and their receptors modulate multiple signalling pathways to regulate neuronal survival and to maintain axonal and dendritic networks and synaptic plasticity. Neurotrophins have potential for the treatment of neurological diseases. However, their therapeutic application has been limited owing to their poor plasma stability, restricted nervous system penetration and, importantly, the pleiotropic actions that derive from their concomitant binding to multiple receptors. One strategy to overcome these limitations is to target individual neurotrophin receptors — such as tropomyosin receptor kinase A (TRKA), TRKB, TRKC, the p75 neurotrophin receptor or sortilin — with small-molecule ligands. Such small molecules might also modulate various aspects of these signalling pathways in ways that are distinct from the programmes triggered by native neurotrophins. By departing from conventional neurotrophin signalling, these ligands might provide novel therapeutic options for a broad range of neurological indications.
Collapse
|
73
|
Manni L, Rocco ML, Bianchi P, Soligo M, Guaragna M, Barbaro SP, Aloe L. Nerve growth factor: basic studies and possible therapeutic applications. Growth Factors 2013; 31:115-22. [PMID: 23777359 DOI: 10.3109/08977194.2013.804073] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The nerve growth factor (NGF) belongs to a family of neurotrophic factors called neurotrophins. It was discovered as a molecule that stimulates the survival and maturation of developing neurons in the peripheral nervous system and has later been shown to protect adult neurons in the degenerating mammalian brain. Basic and clinical studies have been undertaken to use NGF as a therapeutic agent aimed at restoring and maintaining neuronal function in the central nervous system and to determine the mechanisms to safely deliver the molecule into the brain. Recent studies have also recognized that the role of NGF extends far beyond the horizon of nerve cells and even beyond the peripheral and central nervous system. Studies published from our laboratory have shown that topical application of NGF possesses a protective action on human pressure ulcer, corneal ulcer and glaucoma. Here, we will review these studies, supporting the therapeutic potential of NGF.
Collapse
Affiliation(s)
- Luigi Manni
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
74
|
Xia Y, Chen BY, Sun XL, Duan L, Gao GD, Wang JJ, Yung KKL, Chen LW. Presence of proNGF-sortilin signaling complex in nigral dopamine neurons and its variation in relation to aging, lactacystin and 6-OHDA insults. Int J Mol Sci 2013; 14:14085-104. [PMID: 23880857 PMCID: PMC3742233 DOI: 10.3390/ijms140714085] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 06/20/2013] [Accepted: 06/25/2013] [Indexed: 12/15/2022] Open
Abstract
Growing evidence has shown that proNGF-p75NTR-sortilin signaling might be a crucial factor in neurodegeneration, but it remains unclear if it may function in nigral neurons under aging and disease. The purpose of this study is to examine and quantify proNGF and sortilin expression in the substantia nigra and dynamic changes of aging in lactacystin and 6-hydroxydopamine (6-OHDA) rat models of Parkinson’s disease using immunofluorescence, electronic microscopy, western blot and FLIVO staining methods. The expression of proNGF and sortilin was abundantly and selectively identified in tyrosine hydroxylase (TH)-containing dopamine neurons in the substantia nigra. These proNGF/TH, sortilin/TH-positive neurons were densely distributed in the ventral tier, while they were less distributed in the dorsal tier, where calbindin-D28K-containing neurons were numerously located. A correlated decrease of proNGF, sortilin and TH was also detected during animal aging process. While increase of proNGF, sortilin and cleaved (active) caspase-3 expression was found in the lactacystin model, dynamic proNGF and sortilin changes along with dopamine neuronal loss were demonstrated in the substantia nigra of both the lactacystin and 6-OHDA models. This study has thus revealed the presence of the proNGF-sortilin signaling complex in nigral dopamine neurons and its response to aging, lactacystin and 6-OHDA insults, suggesting that it might contribute to neuronal apoptosis or neurodegeneration during pathogenesis and disease progression of Parkinson’s disease; the underlying mechanism and key signaling pathways involved warrant further investigation.
Collapse
Affiliation(s)
- Yi Xia
- Institute of Neurosciences, Fourth Military Medical University, Xi’an 710032, China; E-Mails: (Y.X.); (X.-L.S.); (L.D.)
- Department of Neurosurgery, Tangdou Hospital, Fourth Military Medical University, Xi’an 710038, China; E-Mail:
| | - Bei-Yu Chen
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; E-Mail:
| | - Xiao-Long Sun
- Institute of Neurosciences, Fourth Military Medical University, Xi’an 710032, China; E-Mails: (Y.X.); (X.-L.S.); (L.D.)
| | - Li Duan
- Institute of Neurosciences, Fourth Military Medical University, Xi’an 710032, China; E-Mails: (Y.X.); (X.-L.S.); (L.D.)
| | - Guo-Dong Gao
- Department of Neurosurgery, Tangdou Hospital, Fourth Military Medical University, Xi’an 710038, China; E-Mail:
| | - Jing-Jie Wang
- Department of Gastroenterology, Tangdou Hospital, Fourth Military Medical University, Xi’an 710038, China
- Authors to whom correspondence should be addressed; E-Mails: (J.-J.W.); (L.-W.C.); Tel.: +86-29-8477-6840 (L.-W.C.); Fax: +86-29-8324-6270 (L.-W.C.)
| | - Ken Kam-Lin Yung
- Department of Biology, Baptist University of Hong Kong, Hong Kong, China; E-Mail:
| | - Liang-Wei Chen
- Institute of Neurosciences, Fourth Military Medical University, Xi’an 710032, China; E-Mails: (Y.X.); (X.-L.S.); (L.D.)
- Authors to whom correspondence should be addressed; E-Mails: (J.-J.W.); (L.-W.C.); Tel.: +86-29-8477-6840 (L.-W.C.); Fax: +86-29-8324-6270 (L.-W.C.)
| |
Collapse
|
75
|
Long-term effects of selective immunolesions of cholinergic neurons of the nucleus basalis magnocellularis on the ascending cholinergic pathways in the rat: A model for Alzheimer's disease. Brain Res Bull 2013; 94:9-16. [DOI: 10.1016/j.brainresbull.2013.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Accepted: 01/07/2013] [Indexed: 11/24/2022]
|
76
|
Tirassa P, Maccarone M, Florenzano F, Cartolano S, De Nicolò S. Vascular and neuronal protection induced by the ocular administration of nerve growth factor in diabetic-induced rat encephalopathy. CNS Neurosci Ther 2013; 19:307-18. [PMID: 23528019 DOI: 10.1111/cns.12085] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 01/18/2013] [Accepted: 02/02/2013] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Based on our previous findings on the efficacy of ocular applied nerve growth factor as eye drops (oNGF) to act in brain and counteract neuronal damage, we hypothesized that oNGF treatment might revert neuronal atrophy occurring in diabetic brain also by controlling neurotrophin system changes. The major NGF brain target areas, such as the septum and the hippocampus, were used as an experimental paradigma to test this hypothesis. METHODS Bilateral oNGF treatment was performed twice a day for 2 weeks in full-blown streptozotocin-treated adult male rats. The forebrain distribution of cholinergic and endothelial cell markers and NGF receptors were studied by confocal microscopy. The septo-hippocampal content of NGF mature and precursor form and NGF receptors expression were also analyzed by Elisa and Western blot. RESULTS oNGF treatment recovers the morphological alterations and the neuronal atrophy in septum and normalized the expression of mature and pro-NGF, as well as NGF receptors in the septum and hippocampus of diabetic rats. In addition, oNGF stimulated brain vascularization and up-regulated the TRKA receptor in vessel endothelium. CONCLUSIONS Our findings confirm that reduced availability of mature NGF and NGF signaling impairment favors vascular and neuronal alterations in diabetic septo-hippocampal areas and corroborate the ability of oNGF to act as a neuroprotective agent in brain.
Collapse
Affiliation(s)
- Paola Tirassa
- Institute of Cellular Biology and Neurobiology, National Research Council (CNR), Rome, Italy.
| | | | | | | | | |
Collapse
|
77
|
Ubhi K, Rockenstein E, Vazquez-Roque R, Mante M, Inglis C, Patrick C, Adame A, Fahnestock M, Doppler E, Novak P, Moessler H, Masliah E. Cerebrolysin modulates pronerve growth factor/nerve growth factor ratio and ameliorates the cholinergic deficit in a transgenic model of Alzheimer's disease. J Neurosci Res 2013; 91:167-77. [PMID: 23152192 DOI: 10.1002/jnr.23142] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2012] [Revised: 08/19/2012] [Accepted: 08/21/2012] [Indexed: 11/09/2022]
Abstract
Alzheimer's disease (AD) is characterized by degeneration of neocortex, limbic system, and basal forebrain, accompanied by accumulation of amyloid-β and tangle formation. Cerebrolysin (CBL), a peptide mixture with neurotrophic-like effects, is reported to improve cognition and activities of daily living in patients with AD. Likewise, CBL reduces synaptic and behavioral deficits in transgenic (tg) mice overexpressing the human amyloid precursor protein (hAPP). The neuroprotective effects of CBL may involve multiple mechanisms, including signaling regulation, control of APP metabolism, and expression of neurotrophic factors. We investigate the effects of CBL in the hAPP tg model of AD on levels of neurotrophic factors, including pro-nerve growth factor (NGF), NGF, brain-derived neurotrophic factor (BDNF), neurotropin (NT)-3, NT4, and ciliary neurotrophic factor (CNTF). Immunoblot analysis demonstrated that levels of pro-NGF were increased in saline-treated hAPP tg mice. In contrast, CBL-treated hAPP tg mice showed levels of pro-NGF comparable to control and increased levels of mature NGF. Consistently with these results, immunohistochemical analysis demonstrated increased NGF immunoreactivity in the hippocampus of CBL-treated hAPP tg mice. Protein levels of other neurotrophic factors, including BDNF, NT3, NT4, and CNTF, were unchanged. mRNA levels of NGF and other neurotrophins were also unchanged. Analysis of neurotrophin receptors showed preservation of the levels of TrKA and p75(NTR) immunoreactivity per cell in the nucleus basalis. Cholinergic cells in the nucleus basalis were reduced in the saline-treated hAPP tg mice, and treatment with CBL reduced these cholinergic deficits. These results suggest that the neurotrophic effects of CBL might involve modulation of the pro-NGF/NGF balance and a concomitant protection of cholinergic neurons.
Collapse
Affiliation(s)
- Kiren Ubhi
- Department of Neurosciences, University of California San Diego, La Jolla, California 92093-0624, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Lilja AM, Luo Y, Yu QS, Röjdner J, Li Y, Marini AM, Marutle A, Nordberg A, Greig NH. Neurotrophic and neuroprotective actions of (-)- and (+)-phenserine, candidate drugs for Alzheimer's disease. PLoS One 2013; 8:e54887. [PMID: 23382994 PMCID: PMC3559887 DOI: 10.1371/journal.pone.0054887] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 12/17/2012] [Indexed: 12/11/2022] Open
Abstract
Neuronal dysfunction and demise together with a reduction in neurogenesis are cardinal features of Alzheimer’s disease (AD) induced by a combination of oxidative stress, toxic amyloid-β peptide (Aβ) and a loss of trophic factor support. Amelioration of these was assessed with the Aβ lowering AD experimental drugs (+)-phenserine and (−)-phenserine in neuronal cultures, and actions in mice were evaluated with (+)-phenserine. Both experimental drugs together with the metabolite N1-norphenserine induced neurotrophic actions in human SH-SY5Y cells that were mediated by the protein kinase C (PKC) and extracellular signal–regulated kinases (ERK) pathways, were evident in cells expressing amyloid precursor protein Swedish mutation (APPSWE), and retained in the presence of Aβ and oxidative stress challenge. (+)-Phenserine, together with its (−) enantiomer as well as its N1- and N8-norphenserine and N1,N8-bisnorphenserine metabolites, likewise provided neuroprotective activity against oxidative stress and glutamate toxicity via the PKC and ERK pathways. These neurotrophic and neuroprotective actions were evident in primary cultures of subventricular zone (SVZ) neural progenitor cells, whose neurosphere size and survival were augmented by (+)-phenserine. Translation of these effects in vivo was assessed in wild type and AD APPswe transgenic (Tg2576) mice by doublecortin (DCX) immunohistochemical analysis of neurogenesis in the SVZ, which was significantly elevated by 16 day systemic (+)-phenserine treatment, in the presence of a (+)-phenserine-induced elevation in brain- derived neurotrophic factor (BDNF).
Collapse
Affiliation(s)
- Anna M. Lilja
- Alzheimer Neurobiology Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Drug Design and Development Section, Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
- * E-mail: (AL); (NHG)
| | - Yu Luo
- Department of Neurological Surgery, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Qian-sheng Yu
- Drug Design and Development Section, Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Jennie Röjdner
- Alzheimer Neurobiology Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Yazhou Li
- Drug Design and Development Section, Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Ann M. Marini
- Department of Neurology and Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Amelia Marutle
- Alzheimer Neurobiology Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Agneta Nordberg
- Alzheimer Neurobiology Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Nigel H. Greig
- Drug Design and Development Section, Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
- * E-mail: (AL); (NHG)
| |
Collapse
|
79
|
The olfactory system in Alzheimer’s disease: Pathology, pathophysiology and pathway for therapy. Transl Neurosci 2013. [DOI: 10.2478/s13380-013-0108-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
AbstractOlfaction is frequently mentioned as a “neglected sense”, although the olfactory system has several interesting and unique anatomical and physiological features. Olfactory involvement is present in several degenerative disorders, especially in Alzheimer’s disease (AD). The peripheral and central parts of the olfactory system are damaged even in the early stages of AD, manifesting in profound olfactory deficits. Besides the early pathology, the olfactory system may be involved in the pathogenesis of AD by providing a route of entry for pathological agents still unknown. In contrast to this olfactory vector hypothesis, the olfactory system can be used to deliver therapeutic agents in AD, such as nerve growth factor and insulin, by decreasing the side-effects of the therapy or providing a non-invasive method of delivery.
Collapse
|
80
|
Hardenacke K, Kuhn J, Lenartz D, Maarouf M, Mai JK, Bartsch C, Freund HJ, Sturm V. Stimulate or degenerate: deep brain stimulation of the nucleus basalis Meynert in Alzheimer dementia. World Neurosurg 2012; 80:S27.e35-43. [PMID: 23246738 DOI: 10.1016/j.wneu.2012.12.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 11/16/2012] [Accepted: 12/07/2012] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Deep brain stimulation (DBS) is a therapeutically effective neurosurgical method originally applied in movement disorders. Over time, the application of DBS has increasingly been considered as a therapeutic option for several neuropsychiatric disorders, including Gilles de la Tourette syndrome, obsessive compulsive disorder, major depression and addiction. Latest research suggests beneficial effects of DBS in Alzheimer dementia (AD). Because of the high prevalence and the considerable burden of the disease, we endeavored to discuss and reveal the challenges of DBS in AD. METHODS Recent literature on the pathophysiology of AD, including translational data and human studies, has been studied to generate a fundamental hypothesis regarding the effects of electrical stimulation on cognition and to facilitate our ongoing pilot study regarding DBS of the nucleus basalis Meynert (NBM) in patients with AD. RESULTS It is hypothesized that DBS in the nucleus basalis Meynert could probably improve or at least stabilize memory and cognitive functioning in patients with AD by facilitating neural oscillations and by enhancing the synthesis of nerve growth factors. CONCLUSIONS Considering the large number of patients suffering from AD, there is a great need for novel and effective treatment methods. Our research provides insights into the theoretical background of DBS in AD. Providing that our hypothesis will be validated by our ongoing pilot study, DBS could be an opportunity in the treatment of AD.
Collapse
Affiliation(s)
- Katja Hardenacke
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | | | | | | | | | | | | | | |
Collapse
|
81
|
Nagata T, Shinagawa S, Nukariya K, Nakayama R, Nakayama K, Yamada H. Association between nerve growth factor gene polymorphism and executive dysfunction in Japanese patients with early-stage Alzheimer's disease and amnestic mild cognitive impairment. Dement Geriatr Cogn Disord 2012; 32:379-86. [PMID: 22301435 DOI: 10.1159/000335355] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/15/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS To address the clinical neurocognitive roles of nerve growth factor (NGF) genetic polymorphism in early-stage Alzheimer's disease (AD) and amnestic mild cognitive impairment (A-MCI), we investigated the association between this single-nucleotide polymorphism (SNP) and executive dysfunction as a nonmemory cognitive impairment. METHODS Among 200 outpatients with dementia and MCI whose NGF SNP rs6330 genotype was identified, those with A-MCI (n = 35) and early-stage AD (n = 67) were recruited and divided into three groups according to genotype (C/C: n = 58, C/T: n = 39, T/T: n = 5). Then, the Frontal Assessment Battery (FAB) scores were compared among the three (C/C, C/T, T/T) or two (C/C, T carrier) genotype groups. RESULTS Among the subtests, a significant difference was only noted for the go/no-go scores (p < 0.01) between C/C and T carriers. However, no significant differences in the demographic variables and other neuropsychological subtest scores reflecting attentional and memory function were observed among the genotypes. CONCLUSION Regarding the functional roles of neurotrophin polymorphisms as they relate to executive dysfunction, the NGF gene rs6330 might influence the inhibition task in Japanese patients with early-stage AD or A-MCI.
Collapse
Affiliation(s)
- Tomoyuki Nagata
- Division of Molecular Genetics, Institute of DNA Medicine, Jikei University School of Medicine, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
82
|
Eriksdotter-Jönhagen M, Linderoth B, Lind G, Aladellie L, Almkvist O, Andreasen N, Blennow K, Bogdanovic N, Jelic V, Kadir A, Nordberg A, Sundström E, Wahlund LO, Wall A, Wiberg M, Winblad B, Seiger A, Almqvist P, Wahlberg L. Encapsulated cell biodelivery of nerve growth factor to the Basal forebrain in patients with Alzheimer's disease. Dement Geriatr Cogn Disord 2012; 33:18-28. [PMID: 22377499 DOI: 10.1159/000336051] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/21/2011] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND/AIMS Degeneration of cholinergic neurons in the basal forebrain correlates with cognitive decline in patients with Alzheimer's disease (AD). Targeted delivery of exogenous nerve growth factor (NGF) has emerged as a potential AD therapy due to its regenerative effects on the basal forebrain cholinergic neurons in AD animal models. Here we report the results of a first-in-man study of encapsulated cell (EC) biodelivery of NGF to the basal forebrain of AD patients with the primary objective to explore safety and tolerability. METHODS This was an open-label, 12-month study in 6 AD patients. Patients were implanted stereotactically with EC-NGF biodelivery devices targeting the basal forebrain. Patients were monitored with respect to safety, tolerability, disease progression and implant functionality. RESULTS All patients were implanted successfully with bilateral single or double implants without complications or signs of toxicity. No adverse events were related to NGF or the device. All patients completed the study, including removal of implants at 12 months. Positive findings in cognition, EEG and nicotinic receptor binding in 2 of 6 patients were detected. CONCLUSIONS This study demonstrates that surgical implantation and removal of EC-NGF biodelivery to the basal forebrain in AD patients is safe, well tolerated and feasible.
Collapse
Affiliation(s)
- Maria Eriksdotter-Jönhagen
- Departments of Neurobiology, Caring Sciences and Society, Karolinska Institutet, Stockholm, Sweden. maria.eriksdotter.jonhagen @ ki.se
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Travaglia A, Pietropaolo A, La Mendola D, Nicoletti VG, Rizzarelli E. The inorganic perspectives of neurotrophins and Alzheimer's disease. J Inorg Biochem 2012; 111:130-7. [DOI: 10.1016/j.jinorgbio.2011.10.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 08/30/2011] [Accepted: 10/03/2011] [Indexed: 01/19/2023]
|
84
|
Scott L, Feng J, Kiss T, Needle E, Atchison K, Kawabe TT, Milici AJ, Hajós-Korcsok E, Riddell D, Hajós M. Age-dependent disruption in hippocampal θ oscillation in amyloid-β overproducing transgenic mice. Neurobiol Aging 2012; 33:1481.e13-23. [PMID: 22227005 DOI: 10.1016/j.neurobiolaging.2011.12.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 11/07/2011] [Accepted: 12/05/2011] [Indexed: 01/25/2023]
Abstract
Transgenic mice are used to model increased brain amyloid-β (Aβ) and amyloid plaque formation reflecting Alzheimer's disease pathology. In our study hippocampal network oscillations, population spikes, and long-term potentiation (LTP) were recorded in APPswe/PS1dE9 (APP/PS1) and presenilin1 (PS1) transgenic and wild type mice at 2, 4, and 8 months of age under urethane anesthesia. Hippocampal theta oscillations elicited by brainstem stimulation were similar in wild type and PS1 mice at all age groups. In contrast, APP/PS1 mice showed an age-dependent decrease in hippocampal activity, characterized by a significant decline in elicited theta power and frequency at 4 and 8 months. Magnitudes of population spikes and long-term potentiation in the dentate gyrus were similar across groups at both 4 and 8 months. In APP/PS1 mice, soluble and insoluble Aβ, and hippocampal and cortical plaque load increased with age, and the disruption in hippocampal theta oscillation showed a significant correlation with plaque load. Our study shows that, using in vivo electrophysiological methods, early Aβ-related functional deficits can be robustly detected in the brainstem-hippocampus multisynaptic network.
Collapse
Affiliation(s)
- Liam Scott
- Pfizer Global Research and Development, Groton, CT, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Allen SJ, Watson JJ, Dawbarn D. The neurotrophins and their role in Alzheimer's disease. Curr Neuropharmacol 2011; 9:559-73. [PMID: 22654716 PMCID: PMC3263452 DOI: 10.2174/157015911798376190] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Revised: 08/19/2010] [Accepted: 08/19/2010] [Indexed: 12/15/2022] Open
Abstract
Besides being essential for correct development of the vertebrate nervous system the neurotrophins also play a vital role in adult neuron survival, maintenance and regeneration. In addition they are implicated in the pathogenesis of certain neurodegenerative diseases, and may even provide a therapeutic solution for some. In particular there have been a number of studies on the involvement of nerve growth factor (NGF) and brain derived neurotrophic factor (BDNF) in the development of Alzheimer's disease. This disease is of growing concern as longevity increases worldwide, with little treatment available at the moment to alleviate the condition. Memory loss is one of the earliest symptoms associated with Alzheimer's disease. The brain regions first affected by pathology include the hippocampus, and also the entorhinal cortex and basal cholinergic nuclei which project to the hippocampus; importantly, all these areas are required for memory formation. Both NGF and BDNF are affected early in the disease and this is thought to initiate a cascade of events which exacerbates pathology and leads to the symptoms of dementia. This review briefly describes the pathology, symptoms and molecular processes associated with Alzheimer's disease; it discusses the involvement of the neurotrophins, particularly NGF and BDNF, and their receptors, with changes in BDNF considered particularly in the light of its importance in synaptic plasticity. In addition, the possibilities of neurotrophin-based therapeutics are evaluated.
Collapse
Affiliation(s)
- Shelley J Allen
- Dorothy Hodgkin Building, School of Clinical Sciences, University of Bristol, Bristol BS1 3NY, UK
| | | | | |
Collapse
|
86
|
Stepanichev MY. Current approaches and future directions of gene therapy in Alzheimer’s disease. NEUROCHEM J+ 2011. [DOI: 10.1134/s181971241103010x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
87
|
Niewiadomska G, Mietelska-Porowska A, Mazurkiewicz M. The cholinergic system, nerve growth factor and the cytoskeleton. Behav Brain Res 2011; 221:515-26. [DOI: 10.1016/j.bbr.2010.02.024] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Accepted: 02/10/2010] [Indexed: 01/02/2023]
|
88
|
Enciu AM, Nicolescu MI, Manole CG, Mureşanu DF, Popescu LM, Popescu BO. Neuroregeneration in neurodegenerative disorders. BMC Neurol 2011; 11:75. [PMID: 21699711 PMCID: PMC3146817 DOI: 10.1186/1471-2377-11-75] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2011] [Accepted: 06/23/2011] [Indexed: 02/07/2023] Open
Abstract
Background Neuroregeneration is a relatively recent concept that includes neurogenesis, neuroplasticity, and neurorestoration - implantation of viable cells as a therapeutical approach. Discussion Neurogenesis and neuroplasticity are impaired in brains of patients suffering from Alzheimer's Disease or Parkinson's Disease and correlate with low endogenous protection, as a result of a diminished growth factors expression. However, we hypothesize that the brain possesses, at least in early and medium stages of disease, a "neuroregenerative reserve", that could be exploited by growth factors or stem cells-neurorestoration therapies. Summary In this paper we review the current data regarding all three aspects of neuroregeneration in Alzheimer's Disease and Parkinson's Disease.
Collapse
Affiliation(s)
- Ana M Enciu
- Department of Cellular and Molecular Medicine, School of Medicine, 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | | | | | | | | | | |
Collapse
|
89
|
Ginsberg SD, Mufson EJ, Alldred MJ, Counts SE, Wuu J, Nixon RA, Che S. Upregulation of select rab GTPases in cholinergic basal forebrain neurons in mild cognitive impairment and Alzheimer's disease. J Chem Neuroanat 2011; 42:102-10. [PMID: 21669283 DOI: 10.1016/j.jchemneu.2011.05.012] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 05/26/2011] [Accepted: 05/26/2011] [Indexed: 02/02/2023]
Abstract
Endocytic system dysfunction is one of the earliest disturbances that occur in Alzheimer's disease (AD), and may underlie the selective vulnerability of cholinergic basal forebrain (CBF) neurons during the progression of dementia. Herein we report that genes regulating early and late endosomes are selectively upregulated within CBF neurons in mild cognitive impairment (MCI) and AD. Specifically, upregulation of rab4, rab5, rab7, and rab27 was observed in CBF neurons microdissected from postmortem brains of individuals with MCI and AD compared to age-matched control subjects with no cognitive impairment (NCI). Upregulated expression of rab4, rab5, rab7, and rab27 correlated with antemortem measures of cognitive decline in individuals with MCI and AD. qPCR validated upregulation of these select rab GTPases within microdissected samples of the basal forebrain. Moreover, quantitative immunoblot analysis demonstrated upregulation of rab5 protein expression in the basal forebrain of subjects with MCI and AD. The elevation of rab4, rab5, and rab7 expression is consistent with our recent observations in CA1 pyramidal neurons in MCI and AD. These findings provide further support that endosomal pathology accelerates endocytosis and endosome recycling, which may promote aberrant endosomal signaling and neurodegeneration throughout the progression of AD.
Collapse
Affiliation(s)
- Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY 10962, USA.
| | | | | | | | | | | | | |
Collapse
|
90
|
Barbosa IG, Huguet RB, Neves FS, Reis HJ, Bauer ME, Janka Z, Palotás A, Teixeira AL. Impaired nerve growth factor homeostasis in patients with bipolar disorder. World J Biol Psychiatry 2011; 12:228-32. [PMID: 20923384 DOI: 10.3109/15622975.2010.518629] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Neuro-trophins are critically involved in neuro-plasticity, the impairment of which is a major role-player in bipolar disorder (BD), and their altered levels have been recently advocated in the patho-physiology of this affective malady. The aim of this study, therefore, was to evaluate the plasma levels of nerve growth factor (NGF) in BD patients in comparison with control subjects. METHODS Forty-nine BD type-I individuals (30 in mania and 19 in euthymia) and 36 healthy controls were assessed by Mini-plus, Young mania and Hamilton depression rating scales. NGF levels were detected by ELISA. RESULTS Plasma NGF concentrations were decreased in BD patients when compared to that seen with controls. BD individuals in mania had lower NGF levels than euthymic patients or controls. NGF levels were negatively correlated with the severity of mania. CONCLUSIONS This is the first study to evaluate NGF levels in BD patients, providing further support to the hypothesis of impaired neuro-plasticity in BD. These data also suggest that NGF measurement could be used for the biological marker for manic state.
Collapse
Affiliation(s)
- Izabela Guimarães Barbosa
- Programa de Neurociências, Universidade Federal de Minas Gerais (UFMG), Campus Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
91
|
Croce N, Dinallo V, Ricci V, Federici G, Caltagirone C, Bernardini S, Angelucci F. Neuroprotective effect of neuropeptide Y against β-amyloid 25-35 toxicity in SH-SY5Y neuroblastoma cells is associated with increased neurotrophin production. NEURODEGENER DIS 2011; 8:300-9. [PMID: 21346312 DOI: 10.1159/000323468] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Accepted: 12/09/2010] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND In the central nervous system, several neuropeptides are believed to be involved in the pathophysiology of Alzheimer's disease (AD). Among them, neuropeptide Y (NPY) is a small peptide widely distributed throughout the brain, where it serves as a neurotransmitter and/or a modulator of several neuroendocrine functions. More recently, NPY has generated interest because of its role in neuroprotection against excitotoxicity and modulation of neurogenesis. Interestingly, these effects are also influenced by neurotrophins, critical molecules for the function and survival of neurons that degenerate in AD. OBJECTIVE Our purpose was to investigate whether NPY might be a neuroprotective agent in AD and whether neurotrophins are involved in NPY-induced neuroprotection. METHODS To test this hypothesis, we exposed the SH-SY5Y neuroblastoma cell line to toxic concentrations of β-amyloid (Aβ) peptide fragment 25-35 (Aβ(25-35)) and measured cell survival and neurotrophin expression before and after a preincubation with NPY in the growth medium. RESULTS Our results demonstrated that preincubation with NPY prevented cell loss due to the toxic effect of Aβ(25-35). Moreover, while intracellular production of nerve growth factor and brain-derived neurotrophic factor were reduced by Aβ, NPY restored or even increased neurotrophin protein and mRNA in SH-SY5Y cells. CONCLUSION In conclusion, this study demonstrates that NPY increases the survival of SH-SY5Y neuroblastoma cells and counteracts the toxic effect of Aβ. In addition, NPY restores the neurotrophin levels in these cells. Although preliminary, these observations might be useful to understand the pathology of Alzheimer's and/or develop new therapeutic strategies.
Collapse
|
92
|
Wicklund L, Leão RN, Strömberg AM, Mousavi M, Hovatta O, Nordberg A, Marutle A. Β-amyloid 1-42 oligomers impair function of human embryonic stem cell-derived forebrain cholinergic neurons. PLoS One 2010; 5:e15600. [PMID: 21179413 PMCID: PMC3003688 DOI: 10.1371/journal.pone.0015600] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 11/15/2010] [Indexed: 12/20/2022] Open
Abstract
Cognitive impairment in Alzheimer's disease (AD) patients is associated with a decline in the levels of growth factors, impairment of axonal transport and marked degeneration of basal forebrain cholinergic neurons (BFCNs). Neurogenesis persists in the adult human brain, and the stimulation of regenerative processes in the CNS is an attractive prospect for neuroreplacement therapy in neurodegenerative diseases such as AD. Currently, it is still not clear how the pathophysiological environment in the AD brain affects stem cell biology. Previous studies investigating the effects of the β-amyloid (Aβ) peptide on neurogenesis have been inconclusive, since both neurogenic and neurotoxic effects on progenitor cell populations have been reported. In this study, we treated pluripotent human embryonic stem (hES) cells with nerve growth factor (NGF) as well as with fibrillar and oligomeric Aβ1-40 and Aβ1-42 (nM-µM concentrations) and thereafter studied the differentiation in vitro during 28-35 days. The process applied real time quantitative PCR, immunocytochemistry as well as functional studies of intracellular calcium signaling. Treatment with NGF promoted the differentiation into functionally mature BFCNs. In comparison to untreated cells, oligomeric Aβ1-40 increased the number of functional neurons, whereas oligomeric Aβ1-42 suppressed the number of functional neurons. Interestingly, oligomeric Aβ exposure did not influence the number of hES cell-derived neurons compared with untreated cells, while in contrast fibrillar Aβ1-40 and Aβ1-42 induced gliogenesis. These findings indicate that Aβ1-42 oligomers may impair the function of stem cell-derived neurons. We propose that it may be possible for future AD therapies to promote the maturation of functional stem cell-derived neurons by altering the brain microenvironment with trophic support and by targeting different aggregation forms of Aβ.
Collapse
Affiliation(s)
- Linn Wicklund
- Department of Neurobiology, Care Sciences and Society, Division of Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - Richardson N. Leão
- Department of Neuroscience, Neuronal Oscillation Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Anne-Marie Strömberg
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Malahat Mousavi
- Department of Neurobiology, Care Sciences and Society, Division of Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - Outi Hovatta
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Agneta Nordberg
- Department of Neurobiology, Care Sciences and Society, Division of Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden
- Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Amelia Marutle
- Department of Neurobiology, Care Sciences and Society, Division of Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|