1
|
Kropf E, Shekari A, Jaberi S, Puri A, Wu C, Fahnestock M. Age-induced nitrative stress decreases retrograde transport of proNGF via TrkA and increases proNGF retrograde transport and neurodegeneration via p75 NTR. Front Mol Neurosci 2023; 16:1241420. [PMID: 38025269 PMCID: PMC10679388 DOI: 10.3389/fnmol.2023.1241420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Axonal transport of pro nerve growth factor (proNGF) is impaired in aged basal forebrain cholinergic neurons (BFCNs), which is associated with their degeneration. ProNGF is neurotrophic in the presence of its receptor tropomyosin-related kinase A (TrkA) but induces apoptosis via the pan-neurotrophin receptor (p75NTR) when TrkA is absent. It is well established that TrkA is lost while p75NTR is maintained in aged BFCNs, but whether aging differentially affects transport of proNGF via each receptor is unknown. Nitrative stress increases during aging, but whether age-induced nitrative stress differentially affects proNGF transport via TrkA versus p75NTR has not yet been studied. Answering these questions is essential for developing an accurate understanding of the mechanisms contributing to age-induced loss of proNGF transport and BFCN degeneration. Methods In this study, fluorescence microscopy was used to analyze axonal transport of quantum dot labeled proNGF in rat BFCNs in vitro. Receptor specific effects were studied with proNGF mutants that selectively bind to either TrkA (proNGF-KKE) or p75NTR (proNGF-Δ9-13). Signaling factor activity was quantified via immunostaining. Results Young BFCNs transported proNGF-KKE but not proNGF-Δ9-13, and proNGF transport was not different in p75NTR knockout BFCNs compared to wildtype BFCNs. These results indicate that young BFCNs transport proNGF via TrkA. In vitro aging increased transport of proNGF-Δ9-13 but decreased transport of proNGF-KKE. Treatment with the nitric oxide synthase inhibitor L-NAME reduced retrograde transport of proNGF-Δ9-13 in aged BFCNs while increasing retrograde transport of proNGF-KKE but did not affect TrkA or p75NTR levels. ProNGF-Δ9-13 induced greater pro-apoptotic signaling and neurodegeneration and less pro-survival signaling relative to proNGF-KKE. Discussion Together, these results indicate that age-induced nitrative stress decreases proNGF transport via TrkA while increasing proNGF transport via p75NTR. These transport deficits are associated with decreased survival signaling, increased apoptotic signaling, and neurodegeneration. Our findings elucidate the receptor specificity of age-and nitrative stress-induced proNGF transport deficits. These results may help to rescue the neurotrophic signaling of proNGF in aging to reduce age-induced loss of BFCN function and cognitive decline.
Collapse
Affiliation(s)
- Erika Kropf
- Neuroscience Program, McMaster University, Hamilton, ON, Canada
| | - Arman Shekari
- Neuroscience Program, McMaster University, Hamilton, ON, Canada
| | - Sama Jaberi
- Neuroscience Program, McMaster University, Hamilton, ON, Canada
| | - Anish Puri
- Neuroscience Program, McMaster University, Hamilton, ON, Canada
| | - Chengbiao Wu
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States
| | - Margaret Fahnestock
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
2
|
Arteaga-Silva M, Limón-Morales O, Bonilla-Jaime H, Vigueras-Villaseñor RM, Rojas-Castañeda J, Hernández-Rodríguez J, Montes S, Hernández-González M, Ríos C. Effects of postnatal exposure to cadmium on male sexual incentive motivation and copulatory behavior: Estrogen and androgen receptors expression in adult brain rat. Reprod Toxicol 2023; 120:108445. [PMID: 37482142 DOI: 10.1016/j.reprotox.2023.108445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
There are numerous evidence showing that cadmium (Cd) is an endocrine disruptor that exerts multiple toxic effects at different reproductive levels, including male sexual behavior (MSB). The effect of early exposure to Cd on sexual incentive motivation (SIM) and MSB in adult stage, and the immunoreactivity of receptors for hormones such as estrogens and androgens in brain regions that are relevant for the SIM and MSB display, have not been studied until now. The present study evaluated the effects of 0.5 and 1 mg/kg CdCl2 from day 1-56 of postnatal life on SIM and MSB in adults rats, as well as serum testosterone concentrations, Cd concentration in blood, testis, and brain areas, and the immunoreactivity in estrogen receptors (ER-α and -β), and androgen receptor (AR) in the olfactory bulbs (OB), medial preoptic area (mPOA), and medial amygdala (MeA). Our results showed that both doses of Cd decreased SIM and MSB, accompanied by low serum concentrations of testosterone. Also, there was a significant reduction in immunoreactivity of ER-α and AR in mPOA, and a significant reduction in AR in MeA on male rats treated with Cd 1 mg/kg. These results show that exposure to high doses of Cd in early postnatal life could alter the correct integration of hormonal signals in the brain areas that regulate and display SIM and MSB in adult male rats.
Collapse
Affiliation(s)
- Marcela Arteaga-Silva
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana-Iztapalapa, Av. Ferrocarril San Rafael Atlixco No. 186, Col. Leyes de Reforma 1ª, Sección, Alcaldía Iztapalapa, C.P. 09340, A.P. 55-535, Ciudad de México, México.
| | - Ofelia Limón-Morales
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana-Iztapalapa, Av. Ferrocarril San Rafael Atlixco No. 186, Col. Leyes de Reforma 1ª, Sección, Alcaldía Iztapalapa, C.P. 09340, A.P. 55-535, Ciudad de México, México
| | - Herlinda Bonilla-Jaime
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana-Iztapalapa, Av. Ferrocarril San Rafael Atlixco No. 186, Col. Leyes de Reforma 1ª, Sección, Alcaldía Iztapalapa, C.P. 09340, A.P. 55-535, Ciudad de México, México
| | - Rosa María Vigueras-Villaseñor
- Instituto Nacional de Pediatría, Calzada México Xochimilco No. 101, Colonia San Lorenzo Huipulco, Tlalpan, CP 14370 Ciudad de México, México
| | - Julio Rojas-Castañeda
- Instituto Nacional de Pediatría, Calzada México Xochimilco No. 101, Colonia San Lorenzo Huipulco, Tlalpan, CP 14370 Ciudad de México, México
| | - Joel Hernández-Rodríguez
- Cuerpo Académico de Investigación en Salud de la Licenciatura en Quiropráctica (CA-UNEVE-01), Universidad Estatal del Valle de Ecatepec, Estado de México 55210, México
| | - Sergio Montes
- Unidad Académica Multidisciplinaria Reynosa-Aztlán, Universidad Autónoma de Tamaulipas, Lago de Chapala y Calle 16, Aztlán, Reynosa 88740, México
| | - Marisela Hernández-González
- Instituto de Neurociencias, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Francisco de Quevedo No. 180, Col. Arcos Vallarta, 44130 Guadalajara, Jalisco, México
| | - Camilo Ríos
- Dirección de Investigación, Instituto Nacional de Rehabilitación, Secretaría de Salud, Ciudad de México 14389, México
| |
Collapse
|
3
|
The Nerve Growth Factor Receptor (NGFR/p75 NTR): A Major Player in Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24043200. [PMID: 36834612 PMCID: PMC9965628 DOI: 10.3390/ijms24043200] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
Alzheimer's disease (AD) represents the most prevalent type of dementia in elderly people, primarily characterized by brain accumulation of beta-amyloid (Aβ) peptides, derived from Amyloid Precursor Protein (APP), in the extracellular space (amyloid plaques) and intracellular deposits of the hyperphosphorylated form of the protein tau (p-tau; tangles or neurofibrillary aggregates). The Nerve growth factor receptor (NGFR/p75NTR) represents a low-affinity receptor for all known mammalians neurotrophins (i.e., proNGF, NGF, BDNF, NT-3 e NT-4/5) and it is involved in pathways that determine both survival and death of neurons. Interestingly, also Aβ peptides can blind to NGFR/p75NTR making it the "ideal" candidate in mediating Aβ-induced neuropathology. In addition to pathogenesis and neuropathology, several data indicated that NGFR/p75NTR could play a key role in AD also from a genetic perspective. Other studies suggested that NGFR/p75NTR could represent a good diagnostic tool, as well as a promising therapeutic target for AD. Here, we comprehensively summarize and review the current experimental evidence on this topic.
Collapse
|
4
|
OUP accepted manuscript. Brain 2022; 145:2250-2275. [DOI: 10.1093/brain/awac096] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 11/13/2022] Open
|
5
|
Cuello AC. Rita Levi-Montalcini, NGF Metabolism in Health and in the Alzheimer's Pathology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1331:119-144. [PMID: 34453296 DOI: 10.1007/978-3-030-74046-7_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This chapter relates biographic personal and scientific interactions with Rita Levi-Montalcini. It highlights research from our laboratory inspired by Rita's fundamental discovery. This work from studies on potentially neuro-reparative gangliosides, their interactions with NGF, the role of exogenous NGF in the recovery of degenerating cholinergic neurons of the basal forebrain to the evidence that endogenous NGF maintains the "day-to-day" cortical synaptic phenotype and the discovery of a novel CNS "NGF metabolic pathway." This brain pathway's conceptual platform allowed the investigation of its status during the Alzheimer's disease (AD) pathology. This revealed a major compromise of the conversion of the NGF precursor molecule (proNGF) into the most biologically active molecule, mature NGF (mNGF). Furthermore, in this pathology, we found enhanced protein levels and enzymatic activity of the proteases responsible for the proteolytic degradation of mNGF. A biochemical prospect explaining the tropic factor vulnerability of the NGF-dependent basal forebrain cholinergic neurons and of their synaptic terminals. The NGF deregulation of this metabolic pathway is evident at preclinical stages and reflected in body fluid particularly in the cerebrospinal fluid (CSF). The findings of a deregulation of the NGF metabolic pathway and its reflection in plasma and CSF are opening doors for the development of novel biomarkers for preclinical detection of AD pathology both in Alzheimer's and in Down syndrome (DS) with "silent" AD pathology.
Collapse
Affiliation(s)
- A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.
| |
Collapse
|
6
|
Kropf E, Fahnestock M. Effects of Reactive Oxygen and Nitrogen Species on TrkA Expression and Signalling: Implications for proNGF in Aging and Alzheimer's Disease. Cells 2021; 10:cells10081983. [PMID: 34440751 PMCID: PMC8392605 DOI: 10.3390/cells10081983] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 11/18/2022] Open
Abstract
Nerve growth factor (NGF) and its precursor form, proNGF, are critical for neuronal survival and cognitive function. In the brain, proNGF is the only detectable form of NGF. Dysregulation of proNGF in the brain is implicated in age-related memory loss and Alzheimer’s disease (AD). AD is characterized by early and progressive degeneration of the basal forebrain, an area critical for learning, memory, and attention. Learning and memory deficits in AD are associated with loss of proNGF survival signalling and impaired retrograde transport of proNGF to the basal forebrain. ProNGF transport and signalling may be impaired by the increased reactive oxygen and nitrogen species (ROS/RNS) observed in the aged and AD brain. The current literature suggests that ROS/RNS nitrate proNGF and reduce the expression of the proNGF receptor tropomyosin-related kinase A (TrkA), disrupting its downstream survival signalling. ROS/RNS-induced reductions in TrkA expression reduce cell viability, as proNGF loses its neurotrophic function in the absence of TrkA and instead generates apoptotic signalling via the pan-neurotrophin receptor p75NTR. ROS/RNS also interfere with kinesin and dynein motor functions, causing transport deficits. ROS/RNS-induced deficits in microtubule motor function and TrkA expression and signalling may contribute to the vulnerability of the basal forebrain in AD. Antioxidant treatments may be beneficial in restoring proNGF signalling and axonal transport and reducing basal forebrain neurodegeneration and related deficits in cognitive function.
Collapse
Affiliation(s)
- Erika Kropf
- Graduate Program in Neuroscience, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada;
| | - Margaret Fahnestock
- Department of Psychiatry and Behavioural Neurosciences, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
- Correspondence:
| |
Collapse
|
7
|
Abstract
Due to the growing number of chronic traumatic encephalopathy (CTE) cases in the military and contact sports, defining the cellular and molecular substrate of this disorder is crucial. Most classic neuropathological investigations describe cortical tau and, to a lesser extent, amyloid lesions, which may underlie the clinical sequela associated with CTE. The application of modern molecular biologic technology to postmortem human brain tissue has made it possible to evaluate the genetic signature of specific neuronal phenotypes at different stages of CTE pathology. Most recently, molecular pathobiology has been used in the field of CTE, with an emphasis on the cholinergic neurons located within the nucleus basalis of Meynert, which develop tau pathology and are associated with cognitive dysfunction similar to that found in Alzheimer's disease (AD). Quantitative findings derived from single-cell transcript investigations provide clues to our understanding of the selective vulnerability of neurons containing AD-like tau pathology at different stages of CTE. Since human tissue-based studies provide a gold standard for the field of CTE, continued molecular pathological studies are needed to reveal novel drug targets for the treatment of this disorder.
Collapse
|
8
|
Eftimiadi G, Soligo M, Manni L, Di Giuda D, Calcagni ML, Chiaretti A. Topical delivery of nerve growth factor for treatment of ocular and brain disorders. Neural Regen Res 2021; 16:1740-1750. [PMID: 33510063 PMCID: PMC8328750 DOI: 10.4103/1673-5374.306062] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neurotrophins are a family of proteins that support neuronal proliferation, survival, and differentiation in the central and peripheral nervous systems, and are regulators of neuronal plasticity. Nerve growth factor is one of the best-described neurotrophins and has advanced to clinical trials for treatment of ocular and brain diseases due to its trophic and regenerative properties. Prior trials over the past few decades have produced conflicting results, which have principally been ascribed to adverse effects of systemic nerve growth factor administration, together with poor penetrance of the blood-brain barrier that impairs drug delivery. Contrastingly, recent studies have revealed that topical ocular and intranasal nerve growth factor administration are safe and effective, suggesting that topical nerve growth factor delivery is a potential alternative to both systemic and invasive intracerebral delivery. The therapeutic effects of local nerve growth factor delivery have been extensively investigated for different ophthalmic diseases, including neurotrophic keratitis, glaucoma, retinitis pigmentosa, and dry eye disease. Further, promising pharmacologic effects were reported in an optic glioma model, which indicated that topically administered nerve growth factor diffused far beyond where it was topically applied. These findings support the therapeutic potential of delivering topical nerve growth factor preparations intranasally for acquired and degenerative brain disorders. Preliminary clinical findings in both traumatic and non-traumatic acquired brain injuries are encouraging, especially in pediatric patients, and clinical trials are ongoing. The present review will focus on the therapeutic effects of both ocular and intranasal nerve growth factor delivery for diseases of the brain and eye.
Collapse
Affiliation(s)
- Gemma Eftimiadi
- Institute of Pediatrics, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Marzia Soligo
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Luigi Manni
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Daniela Di Giuda
- Institute of Nuclear Medicine, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Maria Lucia Calcagni
- Institute of Nuclear Medicine, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Antonio Chiaretti
- Institute of Pediatrics, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
9
|
Fetal and Postnatal Nicotine Exposure Modifies Maturation of Gonocytes to Spermatogonia in Mice. ACTA ACUST UNITED AC 2020; 2020:8892217. [PMID: 33381390 PMCID: PMC7758125 DOI: 10.1155/2020/8892217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/23/2020] [Indexed: 12/31/2022]
Abstract
Studies in laboratory animals have shown that male offspring from dams, exposed to nicotine during pregnancy and postnatal periods, show alterations in fertility, although the origin of this is still uncertain. In this study, we examined in a mouse model if the process of gonocyte maturation to spermatogonia was affected in male offspring from dams with nicotine administration during pregnancy and postnatal periods. BALB/C mice, with and without nicotine administrations in pregnancy and postnatal periods, were studied. The animals were euthanized at 3, 7, 10, 16, and 35 days postpartum (dpp). Testicular tissue samples were processed for histological, ultrastructural, and immunohistochemical studies; and testicular lipoperoxidation was determined. It was observed that in the nicotine-exposed animals, there was increased apoptosis and a reduction in the number of gonocytes that matured to spermatogonia. This gonocyte-spermatogonia maturation reduction was associated with a greater immunoreactivity to nicotinic acetylcholine receptors in the germ cells. Lipoperoxidation was similar in both groups until 16 dpp, with significant reduction at 35 dpp. Our findings suggest that nicotine intake during pregnancy and postnatal periods can affect the process of maturation of gonocytes to spermatogonia and the pool of available spermatogonia for spermatogenesis.
Collapse
|
10
|
He B, Perez SE, Lee SH, Ginsberg SD, Malek-Ahmadi M, Mufson EJ. Expression profiling of precuneus layer III cathepsin D-immunopositive pyramidal neurons in mild cognitive impairment and Alzheimer's disease: Evidence for neuronal signaling vulnerability. J Comp Neurol 2020; 528:2748-2766. [PMID: 32323319 PMCID: PMC7492791 DOI: 10.1002/cne.24929] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/06/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022]
Abstract
The precuneus (PreC; Brodmann area 7), a key hub within the default mode network (DMN) displays amyloid and tau-containing neurofibrillary tangle (NFT) pathology during the onset of Alzheimer's disease (AD). PreC layer III projection neurons contain lysosomal hydrolase cathepsin D (CatD), a marker of neurons vulnerable to NFT pathology. Here we applied single population laser capture microdissection coupled with custom-designed microarray profiling to determine the genetic signature of PreC CatD-positive-layer III neurons accrued from postmortem tissue obtained from the Rush Religious Orders Study (RROS) cases with a premortem clinical diagnosis of no cognitive impairment (NCI), mild cognitive impairment (MCI) and AD. Expression profiling revealed significant differential expression of key transcripts in MCI and AD compared to NCI that underlie signaling defects, including dysregulation of genes within the endosomal-lysosomal and autophagy pathways, cytoskeletal elements, AD-related genes, ionotropic and metabotropic glutamate receptors, cholinergic enzymes and receptors, markers of monoamine neurotransmission as well as steroid-related transcripts. Pervasive defects in both MCI and AD were found in select transcripts within these key gene ontology categories, underscoring the vulnerability of these corticocortical projection neurons during the onset and progression of dementia. Select PreC dysregulated genes detected via custom-designed microarray analysis were validated using qPCR. In summary, expression profiling of PreC CatD -positive layer III neurons revealed significant dysregulation of a mosaic of genes in MCI and AD that were not previously appreciated in terms of their indication of systems-wide signaling defects in a key hub of the DMN.
Collapse
Affiliation(s)
- Bin He
- Department of Neurobiology and Neurology, Barrow
Neurological Institute, Phoenix, Arizona
| | - Sylvia E. Perez
- Department of Neurobiology and Neurology, Barrow
Neurological Institute, Phoenix, Arizona
| | - Sang Han Lee
- Center for Biomedical Imaging and Neuromodulation, Nathan
Kline Institute, Orangeburg, New York
- Child and Adolescent Psychiatry, New York University School
of Medicine, New York, New York
| | - Stephen D. Ginsberg
- Center for Dementia Research, Nathan Kline Institute,
Orangeburg, New York
- Department of Psychiatry, New York University School of
Medicine, New York, New York
- Neuroscience & Physiology, New York University School
of Medicine, New York, New York
- NYU Neuroscience Institute, New York University School of
Medicine, New York, New York
| | | | - Elliott J. Mufson
- Department of Neurobiology and Neurology, Barrow
Neurological Institute, Phoenix, Arizona
| |
Collapse
|
11
|
Chen XQ, Mobley WC. Exploring the Pathogenesis of Alzheimer Disease in Basal Forebrain Cholinergic Neurons: Converging Insights From Alternative Hypotheses. Front Neurosci 2019; 13:446. [PMID: 31133787 PMCID: PMC6514132 DOI: 10.3389/fnins.2019.00446] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/18/2019] [Indexed: 01/01/2023] Open
Abstract
Alzheimer disease (AD) represents an oncoming epidemic that without an effective treatment promises to exact extraordinary financial and emotional burdens (Apostolova, 2016). Studies of pathogenesis are essential for defining critical molecular and cellular events and for discovering therapies to prevent or mitigate their effects. Through studies of neuropathology, genetic and cellular, and molecular biology recent decades have provided many important insights. Several hypotheses have been suggested. Documentation in the 1980s of selective loss of cholinergic neurons of the basal forebrain, followed by clinical improvement in those treated with inhibitors of acetylycholinesterase, supported the "cholinergic hypothesis of age-related cognitive dysfunction" (Bartus et al., 1982). A second hypothesis, prompted by the selective loss of cholinergic neurons and the discovery of central nervous system (CNS) neurotrophic factors, including nerve growth factor (NGF), prompted the "deficient neurotrophic hypothesis" (Chen et al., 2018). The most persuasive hypothesis, the amyloid cascade hypothesis first proposed more than 25 years ago (Selkoe and Hardy, 2016), is supported by a wealth of observations. Genetic studies were exceptionally important, pointing to increased dose of the gene for the amyloid precursor protein (APP) in Down syndrome (DS) and a familial AD (FAD) due to duplication of APP and to mutations in APP and in the genes for Presenilin 1 and 2 (PSEN1, 2), which encode the γ-secretase enzyme that processes APP (Dorszewska et al., 2016). The "tau hypothesis" noted the prominence of tau-related pathology and its correlation with dementia (Kametani and Hasegawa, 2018). Recent interest in induction of microglial activation in the AD brain, as well as other manifestations of inflammation, supports the "inflammatory hypothesis" (Mcgeer et al., 2016). We place these findings in the context of the selective, but by no means unique, involvement of BFCNs and their trophic dependence on NGF signaling and speculate as to how pathogenesis in these neurons is initiated, amplified and ultimately results in their dysfunction and death. In so doing we attempt to show how the different hypotheses for AD may interact and reinforce one another. Finally, we address current attempts to prevent and/or treat AD in light of advances in understanding pathogenetic mechanisms and suggest that studies in the DS population may provide unique insights into AD pathogenesis and treatment.
Collapse
Affiliation(s)
- Xu-Qiao Chen
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| | - William C. Mobley
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
12
|
Modulation of the p75 neurotrophin receptor suppresses age-related basal forebrain cholinergic neuron degeneration. Sci Rep 2019; 9:5273. [PMID: 30918278 PMCID: PMC6437186 DOI: 10.1038/s41598-019-41654-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 02/08/2019] [Indexed: 11/19/2022] Open
Abstract
Age-related degeneration of basal forebrain cholinergic neurons (BFCNs) is linked to cognitive impairment. The p75 neurotrophin receptor (p75NTR) has been proposed to mediate neuronal degeneration in aging. Therefore, we tested the hypothesis that modifying p75NTR function would prevent or reverse aging-related neuronal degeneration using LM11A-31, a small molecule p75NTR modulator that downregulates degenerative and upregulates trophic receptor-associated signaling. Morphological analysis in mice showed loss of BFCN area detectable by 18 months of age. Oral administration of LM11A-31 from age 15 to 18 months resulted in a dose-related preservation of BFCN area and one month of treatment from 17 to 18 months also preserved cell area. To evaluate reversal of established neuronal atrophy, animals were treated from 21 to 25 months of age. Treatment was associated with an increase of cell size to a mean area larger than that observed at 18 months, accompanied by increases in mean MS/VDB neurite length, as well as increased cholinergic fiber density and synaptophysin pre-synaptic marker levels in the hippocampus. These findings support the idea that modulation of p75NTR activity can prevent and potentially reverse age-associated BFCN degeneration. Moreover, this may be achieved therapeutically with orally bioavailable agents such as LM11A-31.
Collapse
|
13
|
Fahnestock M, Shekari A. ProNGF and Neurodegeneration in Alzheimer's Disease. Front Neurosci 2019; 13:129. [PMID: 30853882 PMCID: PMC6395390 DOI: 10.3389/fnins.2019.00129] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 02/05/2019] [Indexed: 11/13/2022] Open
Abstract
Profound and early basal forebrain cholinergic neuron (BFCN) degeneration is a hallmark of Alzheimer's disease (AD). Loss of synapses between basal forebrain and hippocampal and cortical target tissue correlates highly with the degree of dementia and is thought to be a major contributor to memory loss. BFCNs depend for their survival, connectivity and function on the neurotrophin nerve growth factor (NGF) which is retrogradely transported from its sites of synthesis in the cortex and hippocampus. The form of NGF found in human brain is proNGF. ProNGF binds to the NGF receptors TrkA and p75NTR, but it binds more strongly to p75NTR and more weakly to TrkA than does mature NGF. This renders proNGF more sensitive to receptor balance than mature NGF. In the healthy brain, where BFCNs express both TrkA and p75NTR, proNGF is neurotrophic, activating TrkA-dependent signaling pathways such as MAPK and Akt-mTOR and eliciting cell survival and neurite outgrowth. However, if TrkA is lost or if p75NTR is increased, proNGF activates p75NTR-dependent apoptotic pathways such as JNK. This receptor sensitivity serves as a neurotrophic/apoptotic switch that eliminates BFCNs that cannot maintain TrkA/p75NTR balance and therefore synaptic connections with their targets. TrkA is increasingly lost in mild cognitive impairment (MCI) and AD. In addition, proNGF accumulates at BFCN terminals in cortex and hippocampus, reducing the amount of trophic factor that reaches BFCN cell bodies. The loss of TrkA and accumulation of proNGF occur early in MCI and correlate with cognitive impairment. Increased levels of proNGF and reduced levels of TrkA lead to BFCN neurodegeneration and eventual p75NTR-dependent apoptosis. In addition, in AD BFCNs suffer from reduced TrkA-dependent retrograde transport which reduces neurotrophic support. Thus, BFCNs are particularly vulnerable to AD due to their dependence upon retrograde trophic support from proNGF signaling and transport.
Collapse
Affiliation(s)
- Margaret Fahnestock
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Arman Shekari
- Neuroscience Graduate Program, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
14
|
Mitra S, Behbahani H, Eriksdotter M. Innovative Therapy for Alzheimer's Disease-With Focus on Biodelivery of NGF. Front Neurosci 2019; 13:38. [PMID: 30804738 PMCID: PMC6370742 DOI: 10.3389/fnins.2019.00038] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/15/2019] [Indexed: 12/31/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder associated with abnormal protein modification, inflammation and memory impairment. Aggregated amyloid beta (Aβ) and phosphorylated tau proteins are medical diagnostic features. Loss of memory in AD has been associated with central cholinergic dysfunction in basal forebrain, from where the cholinergic circuitry projects to cerebral cortex and hippocampus. Various reports link AD progression with declining activity of cholinergic neurons in basal forebrain. The neurotrophic molecule, nerve growth factor (NGF), plays a major role in the maintenance of cholinergic neurons integrity and function, both during development and adulthood. Numerous studies have also shown that NGF contributes to the survival and regeneration of neurons during aging and in age-related diseases such as AD. Changes in neurotrophic signaling pathways are involved in the aging process and contribute to cholinergic and cognitive decline as observed in AD. Further, gradual dysregulation of neurotrophic factors like NGF and brain derived neurotrophic factor (BDNF) have been reported during AD development thus intensifying further research in targeting these factors as disease modifying therapies against AD. Today, there is no cure available for AD and the effects of the symptomatic treatment like cholinesterase inhibitors (ChEIs) and memantine are transient and moderate. Although many AD treatment studies are being carried out, there has not been any breakthrough and new therapies are thus highly needed. Long-term effective therapy for alleviating cognitive impairment is a major unmet need. Discussion and summarizing the new advancements of using NGF as a potential therapeutic implication in AD are important. In summary, the intent of this review is describing available experimental and clinical data related to AD therapy, priming to gain additional facts associated with the importance of NGF for AD treatment, and encapsulated cell biodelivery (ECB) as an efficient tool for NGF delivery.
Collapse
Affiliation(s)
- Sumonto Mitra
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Homira Behbahani
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,Aging Theme, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
15
|
Terada K, Migita K, Matsushima Y, Sugimoto Y, Kamei C, Matsumoto T, Mori M, Matsunaga K, Takata J, Karube Y. Cholinesterase inhibitor rivastigmine enhances nerve growth factor-induced neurite outgrowth in PC12 cells via sigma-1 and sigma-2 receptors. PLoS One 2018; 13:e0209250. [PMID: 30557385 PMCID: PMC6296549 DOI: 10.1371/journal.pone.0209250] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 12/03/2018] [Indexed: 01/27/2023] Open
Abstract
Rivastigmine (Riv) is a potent and selective cholinesterase (acetylcholinesterase, AChE and butyrylcholinesterase, BuChE) inhibitor developed for the treatment of Alzheimer's disease (AD). To elucidate whether Riv causes neuronal differentiation, we examined its effect on nerve growth factor (NGF)-induced neurite outgrowth in PC12 cells. At concentrations of 0-100 μM, Riv was non-toxic in PC12 cells. Riv caused dose-dependent (10-100 μM) enhancement of NGF-induced neurite outgrowth, which was completely inhibited by the TrkA antagonist GW-441756. By contrast, Riv-mediated enhancement of neurite outgrowth was not blocked by the acetylcholine receptor antagonists, scopolamine and hexamethonium. However, the sigma-1 receptor (Sig-1R) antagonist NE-100 and sigma-2 receptor (Sig-2R) antagonist SM-21 each blocked about half of the Riv-mediated enhancement of NGF-induced neurite outgrowth. Interestingly, the simultaneous application of NE-100 and SM-21 completely blocked the enhancement of NGF-induced neurite outgrowth by Riv. These findings suggest that both Sig-1R and Sig-2R play important roles in NGF-induced neurite outgrowth through TrkA and that Riv may contribute to neuronal repair via Sig-1R and Sig-2R in AD therapy.
Collapse
Affiliation(s)
- Kazuki Terada
- Department of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Keisuke Migita
- Department of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Yukari Matsushima
- Faculty of Pharmaceutical Sciences, Yasuda Women’s University, Hiroshima, Japan
| | - Yumi Sugimoto
- Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, Hyogo, Japan
| | - Chiaki Kamei
- Faculty of Pharmaceutical Sciences, Yasuda Women’s University, Hiroshima, Japan
| | - Taichi Matsumoto
- Department of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Masayoshi Mori
- Department of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Kazuhisa Matsunaga
- Department of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Jiro Takata
- Department of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Yoshiharu Karube
- Department of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
16
|
Tiernan CT, Ginsberg SD, He B, Ward SM, Guillozet-Bongaarts AL, Kanaan NM, Mufson EJ, Counts SE. Pretangle pathology within cholinergic nucleus basalis neurons coincides with neurotrophic and neurotransmitter receptor gene dysregulation during the progression of Alzheimer's disease. Neurobiol Dis 2018; 117:125-136. [PMID: 29859871 PMCID: PMC6278831 DOI: 10.1016/j.nbd.2018.05.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 05/30/2018] [Indexed: 01/22/2023] Open
Abstract
Cholinergic basal forebrain neurons of the nucleus basalis of Meynert (nbM) regulate attentional and memory function and are exquisitely prone to tau pathology and neurofibrillary tangle (NFT) formation during the progression of Alzheimer's disease (AD). nbM neurons require the neurotrophin nerve growth factor (NGF), its cognate receptor TrkA, and the pan-neurotrophin receptor p75NTR for their maintenance and survival. Additionally, nbM neuronal activity and cholinergic tone are regulated by the expression of nicotinic (nAChR) and muscarinic (mAChR) acetylcholine receptors as well as receptors modulating glutamatergic and catecholaminergic afferent signaling. To date, the molecular and cellular relationships between the evolution of tau pathology and nbM neuronal survival remain unknown. To address this knowledge gap, we profiled cholinotrophic pathway genes within nbM neurons immunostained for pS422, a pretangle phosphorylation event preceding tau C-terminal truncation at D421, or dual-labeled for pS422 and TauC3, a later stage tau neo-epitope revealed by this same C-terminal truncation event, via single-population custom microarray analysis. nbM neurons were obtained from postmortem tissues from subjects who died with an antemortem clinical diagnosis of no cognitive impairment (NCI), mild cognitive impairment (MCI), or mild/moderate AD. Quantitative analysis revealed significant downregulation of mRNAs encoding TrkA as well as TrkB, TrkC, and the Trk-mediated downstream pro-survival kinase Akt in pS422+ compared to unlabeled, pS422-negative nbM neurons. In addition, pS422+ neurons displayed a downregulation of transcripts encoding NMDA receptor subunit 2B, metabotropic glutamate receptor 2, D2 dopamine receptor, and β1 adrenoceptor. By contrast, transcripts encoding p75NTR were downregulated in dual-labeled pS422+/TauC3+ neurons. Appearance of the TauC3 epitope was also associated with an upregulation of the α7 nAChR subunit and differential downregulation of the β2 nAChR subunit. Notably, we found that gene expression patterns for each cell phenotype did not differ with clinical diagnosis. However, linear regression revealed that global cognition and Braak stage were predictors of select transcript changes within both unlabeled and pS422+/TauC3- neurons. Taken together, these cell phenotype-specific gene expression profiling data suggest that dysregulation of neurotrophic and neurotransmitter signaling is an early pathogenic mechanism associated with NFT formation in vulnerable nbM neurons and cognitive decline in AD, which may be amenable to therapeutic intervention early in the disease process.
Collapse
Affiliation(s)
- Chelsea T Tiernan
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; Department of Psychiatry, NYU Langone School of Medicine, New York, NY, USA; Department of Physiology & Neuroscience, NYU Langone School of Medicine, New York, NY, USA; NYU Neuroscience Institute, NYU Langone School of Medicine, New York, NY, USA
| | - Bin He
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Sarah M Ward
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | | | - Nicholas M Kanaan
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA; Hauenstein Neurosciences Center, Mercy Health Saint Mary's Hospital, Grand Rapids, MI, USA
| | - Elliott J Mufson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Scott E Counts
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA; Hauenstein Neurosciences Center, Mercy Health Saint Mary's Hospital, Grand Rapids, MI, USA; Department of Family Medicine, Michigan State University, Grand Rapids, MI, USA; Michigan Alzheimer's Disease Core Center, Ann Arbor, MI, USA.
| |
Collapse
|
17
|
Allard S, Jacobs ML, Do Carmo S, Cuello AC. Compromise of cortical proNGF maturation causes selective retrograde atrophy in cholinergic nucleus basalis neurons. Neurobiol Aging 2018; 67:10-20. [PMID: 29609077 DOI: 10.1016/j.neurobiolaging.2018.03.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 02/13/2018] [Accepted: 03/01/2018] [Indexed: 12/19/2022]
Abstract
The degeneration of basal forebrain cholinergic neurons (BFCNs) in Alzheimer's disease (AD) contributes to cognitive impairment. Nerve growth factor (NGF) secreted in the cerebral cortex is necessary for the phenotypic maintenance of BFCNs. AD is associated with disturbances in NGF metabolism, leading to reduced mature NGF levels and to an accumulation of its precursor, proNGF. We previously described that, in rats, this neurotrophic imbalance is sufficient to induce a loss of cortical cholinergic synapses. In the present study, we investigated whether this neurotrophic imbalance can produce an AD-like retrograde degeneration of BFCNs. Using a combination of retrograde labeling and quantitative cell imaging, we could demonstrate that inhibiting cortical proNGF maturation results in an atrophy of BFCNs, a downregulation of the NGF receptors p75 neurotrophin receptor and tropomyosin receptor kinase A, and a reduction in choline acetyltransferase protein expression. The transient increase in sortilin levels and the sustained colocalization with p75 neurotrophin receptor suggest a participation of proNGF in this degenerative process. This study demonstrates that impairments in the extracellular maturation of proNGF are sufficient to cause a somatodendritic retrograde degeneration of the BFCNs.
Collapse
Affiliation(s)
- Simon Allard
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Marie L Jacobs
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Sonia Do Carmo
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
18
|
Tiernan CT, Mufson EJ, Kanaan NM, Counts SE. Tau Oligomer Pathology in Nucleus Basalis Neurons During the Progression of Alzheimer Disease. J Neuropathol Exp Neurol 2018; 77:246-259. [PMID: 29378005 PMCID: PMC6251641 DOI: 10.1093/jnen/nlx120] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Although tau is the primary constituent of neurofibrillary tangles (NFTs), evidence suggests that its toxic moiety is oligomeric in Alzheimer disease (AD). In this regard, tau oligomers correlate more strongly with neuronal loss than NFTs and exhibit neurotoxicity in preclinical AD models. Here, we investigated the spatiotemporal progression of oligomeric tau accumulation within the highly vulnerable cholinergic neurons of the nucleus basalis of Meynert (nbM) in AD. Tissue from subjects who died with a clinical diagnosis of no cognitive impairment, mild cognitive impairment, or AD was immunostained with the tau oligomeric complex 1 (TOC1) antibody, a marker of tau oligomers, and p75NTR, a cholinergic cell marker. Stereological estimates revealed a significant increase in the number of TOC1 nbM immunopositive (+) neurons with a concomitant decrease in p75NTR+ nbM neurons during the transition from mild cognitive impairment to AD. Immunofluorescence identified TOC1+ neurons that colocalized with the pretangle tau marker phospho-Ser422, which persisted into late stage NFTs immunoreactive for MN423. Analysis of the nbM subfields revealed a topographical caudal to rostral gradient of TOC1+ neurons during disease progression. Taken together, these data suggest that toxic tau oligomers accumulate caudorostrally in selectively vulnerable nbM neurons during the onset of AD.
Collapse
Affiliation(s)
| | - Elliott J Mufson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, Arizona
| | - Nicholas M Kanaan
- Department of Translational Science and Molecular Medicine
- Mercy Health Saint Mary’s Hospital, Hauenstein Neurosciences Center, Grand Rapids
| | - Scott E Counts
- Department of Translational Science and Molecular Medicine
- Department of Family Medicine, Michigan State University, Grand Rapids, Michigan
- Mercy Health Saint Mary’s Hospital, Hauenstein Neurosciences Center, Grand Rapids
- Michigan Alzheimer’s Disease Core Center, Ann Arbor, Michigan
| |
Collapse
|
19
|
Pramanik S, Sulistio YA, Heese K. Neurotrophin Signaling and Stem Cells-Implications for Neurodegenerative Diseases and Stem Cell Therapy. Mol Neurobiol 2016; 54:7401-7459. [PMID: 27815842 DOI: 10.1007/s12035-016-0214-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 10/11/2016] [Indexed: 02/07/2023]
Abstract
Neurotrophins (NTs) are members of a neuronal growth factor protein family whose action is mediated by the tropomyosin receptor kinase (TRK) receptor family receptors and the p75 NT receptor (p75NTR), a member of the tumor necrosis factor (TNF) receptor family. Although NTs were first discovered in neurons, recent studies have suggested that NTs and their receptors are expressed in various types of stem cells mediating pivotal signaling events in stem cell biology. The concept of stem cell therapy has already attracted much attention as a potential strategy for the treatment of neurodegenerative diseases (NDs). Strikingly, NTs, proNTs, and their receptors are gaining interest as key regulators of stem cells differentiation, survival, self-renewal, plasticity, and migration. In this review, we elaborate the recent progress in understanding of NTs and their action on various stem cells. First, we provide current knowledge of NTs, proNTs, and their receptor isoforms and signaling pathways. Subsequently, we describe recent advances in the understanding of NT activities in various stem cells and their role in NDs, particularly Alzheimer's disease (AD) and Parkinson's disease (PD). Finally, we compile the implications of NTs and stem cells from a clinical perspective and discuss the challenges with regard to transplantation therapy for treatment of AD and PD.
Collapse
Affiliation(s)
- Subrata Pramanik
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 133-791, Republic of Korea
| | - Yanuar Alan Sulistio
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 133-791, Republic of Korea
| | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 133-791, Republic of Korea.
| |
Collapse
|
20
|
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
|
21
|
Campos C, Rocha NBF, Lattari E, Paes F, Nardi AE, Machado S. Exercise-induced neuroprotective effects on neurodegenerative diseases: the key role of trophic factors. Expert Rev Neurother 2016; 16:723-34. [PMID: 27086703 DOI: 10.1080/14737175.2016.1179582] [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] [Indexed: 01/08/2023]
Abstract
Age-related neurodegenerative disorders, like Alzheimer's or Parkinson's disease, are becoming a major issue to public health care. Currently, there is no effective pharmacological treatment to address cognitive impairment in these patients. Here, we aim to explore the role of exercise-induced trophic factor enhancement in the prevention or delay of cognitive decline in patients with neurodegenerative diseases. There is a significant amount of evidence from animal and human studies that links neurodegenerative related cognitive deficits with changes on brain and peripheral trophic factor levels. Several trials with elderly individuals and patients with neurodegenerative diseases report exercise induced cognitive improvements and changes on trophic factor levels including BDNF, IGF-I, among others. Further studies with healthy aging and clinical populations are needed to understand how diverse exercise interventions produce different variations in trophic factor signaling. Genetic profiles and potential confounders regarding trophic factors should also be addressed in future trials.
Collapse
Affiliation(s)
- Carlos Campos
- a Laboratory of Panic and Respiration , Institute of Psychiatry of Federal University of Rio de Janeiro (IPUB/UFRJ) , Rio de Janeiro , Brazil.,b School of Allied Health Sciences , Polytechnic Institute of Porto , Porto , Portugal
| | - Nuno Barbosa F Rocha
- b School of Allied Health Sciences , Polytechnic Institute of Porto , Porto , Portugal
| | - Eduardo Lattari
- a Laboratory of Panic and Respiration , Institute of Psychiatry of Federal University of Rio de Janeiro (IPUB/UFRJ) , Rio de Janeiro , Brazil
| | - Flávia Paes
- a Laboratory of Panic and Respiration , Institute of Psychiatry of Federal University of Rio de Janeiro (IPUB/UFRJ) , Rio de Janeiro , Brazil
| | - António E Nardi
- a Laboratory of Panic and Respiration , Institute of Psychiatry of Federal University of Rio de Janeiro (IPUB/UFRJ) , Rio de Janeiro , Brazil
| | - Sérgio Machado
- a Laboratory of Panic and Respiration , Institute of Psychiatry of Federal University of Rio de Janeiro (IPUB/UFRJ) , Rio de Janeiro , Brazil.,c Physical Activity Neuroscience Laboratory , Physical Activity Sciences Postgraduate Program - Salgado de Oliveira University (UNIVERSO) , Niterói , Brazil
| |
Collapse
|
22
|
Rojas-Castañeda JC, Vigueras-Villaseñor RM, Chávez-Saldaña M, Rojas P, Gutiérrez-Pérez O, Rojas C, Arteaga-Silva M. Neonatal exposure to monosodium glutamate induces morphological alterations in suprachiasmatic nucleus of adult rat. Int J Exp Pathol 2016; 97:18-26. [PMID: 26799547 PMCID: PMC4840248 DOI: 10.1111/iep.12157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 10/07/2015] [Indexed: 01/19/2023] Open
Abstract
Neonatal exposure to monosodium glutamate (MSG) induces circadian disorders in several physiological and behavioural processes regulated by the suprachiasmatic nucleus (SCN). The objective of this study was to evaluate the effects of neonatal exposure to MSG on locomotor activity, and on morphology, cellular density and expression of proteins, as evaluated by optical density (OD), of vasopressin (VP)-, vasoactive intestinal polypeptide (VIP)- and glial fibrillary acidic protein (GFAP)-immunoreactive cells in the SCN. Male Wistar rats were used: the MSG group was subcutaneously treated from 3 to 10 days of age with 3.5 mg/g/day. Locomotor activity was evaluated at 90 days of age using 'open-field' test, and the brains were processed for immunohistochemical studies. MSG exposure induced a significant decrease in locomotor activity. VP- and VIP-immunoreactive neuronal densities showed a significant decrease, while the somatic OD showed an increase. Major axes and somatic area were significantly increased in VIP neurons. The cellular and optical densities of GFAP-immunoreactive sections of SCN were significantly increased. These results demonstrated that newborn exposure to MSG induced morphological alterations in SCN cells, an alteration that could be the basis for behavioural disorders observed in the animals.
Collapse
Affiliation(s)
| | - Rosa María Vigueras-Villaseñor
- Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, México D.F., México
- Departamento de Morfología, Facultad de Medicina Veterinaria y Zootécnia, UNAM, México D.F., México
| | | | - Patricia Rojas
- Laboratorio de Neurotoxicología, Instituto Nacional de Neurología y Neurocirugía, 'Manuel Velasco Suárez', México D.F., México
| | - Oscar Gutiérrez-Pérez
- Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, México D.F., México
- Departamento de Morfología, Facultad de Medicina Veterinaria y Zootécnia, UNAM, México D.F., México
| | - Carolina Rojas
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, UNAM, México D.F., México
| | | |
Collapse
|
23
|
Hou XQ, Zhang L, Yang C, Rong CP, He WQ, Zhang CX, Li S, Su RY, Chang X, Qin JH, Chen YB, Xian SX, Wang Q. Alleviating effects of Bushen-Yizhi formula on ibotenic acid-induced cholinergic impairments in rat. Rejuvenation Res 2016; 18:111-27. [PMID: 25482164 DOI: 10.1089/rej.2014.1603] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
This study explored the curative effect and underlying mechanisms of a traditional Chinese medicine compound prescription, Bushen-Yizhi formula (BSYZ), in ibotenic acid (IBO)-induced rats. Morris water maze and novel object recognition tests showed that BSYZ significantly improved spatial and object memory. Brain immunohistochemistry staining showed that BSYZ significantly up-regulated expression of choline acetyltransferase (ChAT) and nerve growth factor (NGF) in the hippocampus and cortex. The protein tyrosine kinase high-affinity receptor TrkA was slightly increased in the hippocampus and cortex, and significantly enhanced in the nucleus basalis of Meynert (NBM) after BSYZ intervention. The immunoreactivity of the p75 low-affinity receptor in BSYZ-treated rats was significantly strengthened in the cortex. Similar expression trends of nerve growth factor (NGF), TrkA, and p75 mRNA were observed in the hippocampus and cortex. Additionally, BSYZ reversed IBO-induced disorders of acetylcholine (ACh) levels, ChAT, and cholinesterase (ChE) in the cortex, which was consistent with the changes in mRNA levels of ChAT and acetylcholinesterase (AChE). Expression of ChAT and AChE proteins and mRNA in the hippocampus was up-regulated, whereas the apoptosis-relative protein cleaved caspase-3 was decreased after administration of BSYZ. Moreover, changes in cell death were confirmed by histological morphology. Thus, the results indicated that the BSYZ formula could ameliorate memory impairments in IBO-induced rats, and it exerted its therapeutic action probably by modulating cholinergic pathways, NGF signaling, and anti-apoptosis. Overall, it is suggested that the BSYZ formula might be a potential therapeutic approach for the treatment of Alzheimer's disease (AD) and other cholinergic impairment-related diseases.
Collapse
Affiliation(s)
- Xue-Qin Hou
- 1 DME Center, Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine , Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Barage SH, Sonawane KD. Amyloid cascade hypothesis: Pathogenesis and therapeutic strategies in Alzheimer's disease. Neuropeptides 2015; 52:1-18. [PMID: 26149638 DOI: 10.1016/j.npep.2015.06.008] [Citation(s) in RCA: 372] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 06/24/2015] [Accepted: 06/24/2015] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease is an irreversible, progressive neurodegenerative disorder. Various therapeutic approaches are being used to improve the cholinergic neurotransmission, but their role in AD pathogenesis is still unknown. Although, an increase in tau protein concentration in CSF has been described in AD, but several issues remains unclear. Extensive and accurate analysis of CSF could be helpful to define presence of tau proteins in physiological conditions, or released during the progression of neurodegenerative disease. The amyloid cascade hypothesis postulates that the neurodegeneration in AD caused by abnormal accumulation of amyloid beta (Aβ) plaques in various areas of the brain. The amyloid hypothesis has continued to gain support over the last two decades, particularly from genetic studies. Therefore, current research progress in several areas of therapies shall provide an effective treatment to cure this devastating disease. This review critically evaluates general biochemical and physiological functions of Aβ directed therapeutics and their relevance.
Collapse
Affiliation(s)
- Sagar H Barage
- Department of Biotechnology, Shivaji University, Kolhapur 416004, Maharashtra (M.S.), India
| | - Kailas D Sonawane
- Structural Bioinformatics Unit, Department of Biochemistry, Shivaji University, Kolhapur 416004, Maharashtra (M.S.), India; Department of Microbiology, Shivaji University, Kolhapur 416004, Maharashtra (M.S.), India.
| |
Collapse
|
25
|
Pivtoraiko VN, Abrahamson EE, Leurgans SE, DeKosky ST, Mufson EJ, Ikonomovic MD. Cortical pyroglutamate amyloid-β levels and cognitive decline in Alzheimer's disease. Neurobiol Aging 2015; 36:12-9. [PMID: 25048160 PMCID: PMC4268150 DOI: 10.1016/j.neurobiolaging.2014.06.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/12/2014] [Accepted: 06/19/2014] [Indexed: 02/02/2023]
Abstract
Posterior cingulate cortex (PCC) accumulates amyloid-β (Aβ) early in Alzheimer's disease (AD). The relative concentrations of full-length Aβ and truncated, pyroglutamate-modified Aβ (NpE3) forms, and their correlations to cognitive dysfunction in AD, are unknown. We quantified AβNpE3-42, AβNpE3-40, Aβ1-42, and Aβ1-40 concentrations in soluble (nonfibrillar) and insoluble (fibrillar) pools in PCC from subjects with an antemortem clinical diagnosis of no cognitive impairment, mild cognitive impairment, or mild-moderate AD. In clinical AD, increased PCC concentrations of Aβ were observed for all Aβ forms in the insoluble pool but only for Aβ1-42 in the soluble pool. Lower Mini-Mental State Exam and episodic memory scores correlated most strongly with higher concentrations of soluble and insoluble Aβ1-42. Greater neuropathology severity by Consortium to Establish a Registry for Alzheimer's Disease and National Institute on Aging-Reagan pathologic criteria was associated with higher concentrations of all measured Aβ forms, except soluble AβNpE3-40. Low concentrations of soluble pyroglutamate Aβ across clinical groups likely reflect its rapid sequestration into plaques, thus, the conversion to fibrillar Aβ may be a therapeutic target.
Collapse
Affiliation(s)
| | - Eric E Abrahamson
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA; Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Sue E Leurgans
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL
| | - Steven T DeKosky
- Department of Neurology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Elliott J Mufson
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Milos D Ikonomovic
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA; Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
26
|
Gotanda Y, Wei FY, Harada H, Ohta K, Nakamura KI, Tomizawa K, Ushijima K. Efficient transduction of 11 poly-arginine peptide in an ischemic lesion of mouse brain. J Stroke Cerebrovasc Dis 2014; 23:2023-2030. [PMID: 25081308 DOI: 10.1016/j.jstrokecerebrovasdis.2014.02.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 02/08/2014] [Accepted: 02/26/2014] [Indexed: 11/25/2022] Open
Abstract
Direct intracellular delivery of intact proteins has been successfully achieved by tagging cell-penetrating peptide (CPP), which consists of short positively charged amino acids, such as 11 poly-arginine (11R); however, in vivo delivery of the proteins to the brain has remained challenging because it is unclear whether CPP would enable proteins to cross the blood-brain barrier (BBB). In this study, we conducted an in vivo kinetic study to investigate the efficiency of 11R-mediated peptide delivery in the normal and ischemic brain. The 11R was observed in the microvessels and neurons surrounding the microvessels throughout the brain 1 hour after systemic administration, but the signal of the peptide was faint after 2 hours. In a transient middle cerebral artery occlusion mouse model, 11R was markedly enhanced and remained detectable in the cells on the ipsilateral side for as long as 8 hours after administration compared with the contralateral side. These results suggest that 11R is capable of in vivo delivery to the brain by passing through the BBB. Furthermore, 11R-mediated protein transduction could be used for the delivery of therapeutic molecules in cerebral ischemia.
Collapse
Affiliation(s)
- Yuki Gotanda
- Department of Anesthesiology, Kurume University School of Medicine, Fukuoka, Japan
| | - Fan-Yan Wei
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideki Harada
- Department of Anesthesiology, Kurume University School of Medicine, Fukuoka, Japan
| | - Keisuke Ohta
- Division of Microscopic and Developmental Anatomy, Department of Anatomy, Kurume University School of Medicine, Fukuoka, Japan
| | - Kei-Ichiro Nakamura
- Division of Microscopic and Developmental Anatomy, Department of Anatomy, Kurume University School of Medicine, Fukuoka, Japan
| | - Kazuhito Tomizawa
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| | - Kazuo Ushijima
- Department of Anesthesiology, Kurume University School of Medicine, Fukuoka, Japan
| |
Collapse
|
27
|
Iulita MF, Cuello AC. Nerve growth factor metabolic dysfunction in Alzheimer's disease and Down syndrome. Trends Pharmacol Sci 2014; 35:338-48. [PMID: 24962069 DOI: 10.1016/j.tips.2014.04.010] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 04/16/2014] [Accepted: 04/30/2014] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative condition and the most common type of amnestic dementia in the elderly. Individuals with Down syndrome (DS) are at increased risk of developing AD in adulthood as a result of chromosome 21 trisomy and triplication of the amyloid precursor protein (APP) gene. In both conditions, the central nervous system (CNS) basal forebrain cholinergic system progressively degenerates, and such changes contribute to the manifestation of cognitive decline and dementia. Given the strong dependency of these neurons on nerve growth factor (NGF), it was hypothesized that their atrophy was caused by NGF deficits. However, in AD, the synthesis of NGF is not affected at the transcript level and there is a marked increase in its precursor, proNGF. This apparent paradox remained elusive for many years. In this review, we discuss the recent evidence supporting a CNS deficit in the extracellular metabolism of NGF, both in AD and in DS brains. We describe the nature of this trophic disconnection and its implication for the atrophy of basal forebrain cholinergic neurons. We further discuss the potential of NGF pathway markers as diagnostic indicators of a CNS trophic disconnection.
Collapse
Affiliation(s)
- M Florencia Iulita
- Department of Pharmacology and Therapeutics, McGill University, Montreal, H3G1Y6, Canada
| | - A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, Montreal, H3G1Y6, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, H3G1Y6, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, H3G1Y6, Canada.
| |
Collapse
|
28
|
George S, Rönnbäck A, Gouras GK, Petit GH, Grueninger F, Winblad B, Graff C, Brundin P. Lesion of the subiculum reduces the spread of amyloid beta pathology to interconnected brain regions in a mouse model of Alzheimer's disease. Acta Neuropathol Commun 2014; 2:17. [PMID: 24517102 PMCID: PMC3932948 DOI: 10.1186/2051-5960-2-17] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 02/03/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The progressive development of Alzheimer's disease (AD) pathology follows a spatiotemporal pattern in the human brain. In a transgenic (Tg) mouse model of AD expressing amyloid precursor protein (APP) with the arctic (E693G) mutation, pathology spreads along anatomically connected structures. Amyloid-β (Aβ) pathology first appears in the subiculum and is later detected in interconnected brain regions, including the retrosplenial cortex. We investigated whether the spatiotemporal pattern of Aβ pathology in the Tg APP arctic mice to interconnected brain structures can be interrupted by destroying neurons using a neurotoxin and thereby disconnecting the neural circuitry. RESULTS We performed partial unilateral ibotenic acid lesions of the subiculum (first structure affected by Aβ pathology) in young Tg APParc mice, prior to the onset of pathology. We assessed Aβ/C99 pathology in mice aged up to 6 months after injecting ibotenate into the subiculum. Compared to the brains of intact Tg APP arctic mice, we observed significantly decreased Aβ/C99 pathology in the ipsilateral dorsal subiculum, CA1 region of the hippocampus and the retrosplenial cortex; regions connecting to and from the dorsal subiculum. By contrast, Aβ/C99 pathology was unchanged in the contralateral hippocampus in the mice with lesions. CONCLUSION These results, obtained in an animal model of AD, support the notion that Aβ/C99 pathology is transmitted between interconnected neurons in AD.
Collapse
Affiliation(s)
- Sonia George
- Neuronal Survival Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Annica Rönnbäck
- Alzheimer Disease Research Center, Karolinska Institutet, Stockholm, Sweden
| | - Gunnar K Gouras
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Géraldine H Petit
- Neuronal Survival Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Fiona Grueninger
- CNS Discovery and Translation Pharma Research and Exploratory Development, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Bengt Winblad
- Alzheimer Disease Research Center, Karolinska Institutet, Stockholm, Sweden
| | - Caroline Graff
- Alzheimer Disease Research Center, Karolinska Institutet, Stockholm, Sweden
- Department of Geriatric Medicine, Genetics unit, Karolinska University Hospital, Stockholm, Sweden
| | - Patrik Brundin
- Neuronal Survival Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA
| |
Collapse
|
29
|
Perez SE, Raghanti MA, Hof PR, Kramer L, Ikonomovic MD, Lacor PN, Erwin JM, Sherwood CC, Mufson EJ. Alzheimer's disease pathology in the neocortex and hippocampus of the western lowland gorilla (Gorilla gorilla gorilla). J Comp Neurol 2013; 521:4318-38. [PMID: 23881733 PMCID: PMC6317365 DOI: 10.1002/cne.23428] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 06/28/2013] [Accepted: 07/10/2013] [Indexed: 12/11/2022]
Abstract
The two major histopathologic hallmarks of Alzheimer's disease (AD) are amyloid beta protein (Aβ) plaques and neurofibrillary tangles (NFT). Aβ pathology is a common feature in the aged nonhuman primate brain, whereas NFT are found almost exclusively in humans. Few studies have examined AD-related pathology in great apes, which are the closest phylogenetic relatives of humans. In the present study, we examined Aβ and tau-like lesions in the neocortex and hippocampus of aged male and female western lowland gorillas using immunohistochemistry and histochemistry. Analysis revealed an age-related increase in Aβ-immunoreactive plaques and vasculature in the gorilla brain. Aβ plaques were more abundant in the neocortex and hippocampus of females, whereas Aβ-positive blood vessels were more widespread in male gorillas. Plaques were also Aβ40-, Aβ42-, and Aβ oligomer-immunoreactive, but only weakly thioflavine S- or 6-CN-PiB-positive in both sexes, indicative of the less fibrillar (diffuse) nature of Aβ plaques in gorillas. Although phosphorylated neurofilament immunostaining revealed a few dystrophic neurites and neurons, choline acetyltransferase-immunoreactive fibers were not dystrophic. Neurons stained for the tau marker Alz50 were found in the neocortex and hippocampus of gorillas at all ages. Occasional Alz50-, MC1-, and AT8-immunoreactive astrocyte and oligodendrocyte coiled bodies and neuritic clusters were seen in the neocortex and hippocampus of the oldest gorillas. This study demonstrates the spontaneous presence of both Aβ plaques and tau-like lesions in the neocortex and hippocampus in old male and female western lowland gorillas, placing this species at relevance in the context of AD research.
Collapse
Affiliation(s)
| | - Mary Ann Raghanti
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, Ohio 44242
- Cleveland Metroparks Zoo, Cleveland, Ohio 44109
| | - Patrick R. Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | | | - Milos D. Ikonomovic
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pennsylvania 15213
- Departments of Neurology and Psychiatry, University of Pittsburgh, Pennsylvania 15213
| | - Pascale N. Lacor
- Neurobiology Department and Cognitive Neurology and Alzheimer’s Disease Center, Northwestern University, Evanston, Illinois 60208
| | - Joseph M. Erwin
- Department of Anthropology, The George Washington University, Washington, DC 20052
| | - Chet C. Sherwood
- Department of Anthropology, The George Washington University, Washington, DC 20052
| | | |
Collapse
|
30
|
Knowles JK, Simmons DA, Nguyen TVV, Vander Griend L, Xie Y, Zhang H, Yang T, Pollak J, Chang T, Arancio O, Buckwalter MS, Wyss-Coray T, Massa SM, Longo FM. Small molecule p75NTR ligand prevents cognitive deficits and neurite degeneration in an Alzheimer's mouse model. Neurobiol Aging 2013; 34:2052-63. [PMID: 23545424 PMCID: PMC9035212 DOI: 10.1016/j.neurobiolaging.2013.02.015] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 02/11/2013] [Accepted: 02/18/2013] [Indexed: 01/03/2023]
Abstract
The p75 neurotrophin receptor (p75(NTR)) is associated with multiple mechanisms linked to Alzheimer's disease (AD); hence, modulating its function might confer therapeutic effects. In previous in vitro work, we developed small molecule p75(NTR) ligands that inhibited amyloid-β-induced degenerative signaling and prevented neurite degeneration. In the present study, a prototype p75(NTR) ligand, LM11A-31, was administered orally to the Thy-1 hAPP(Lond/Swe) (APP(L/S)) AD mouse model. LM11A-31 reached brain concentrations known to inhibit degenerative signaling without toxicity or induction of hyperalgesia. It prevented deficits in novel object recognition after 2.5 months and, in a separate cohort, deficits in Y-maze performance after 3 months of treatment. Stereology studies found that the number and size of basal forebrain cholinergic neurons, which are normal in APP(L/S) mice, were unaffected. Neuritic dystrophy, however, was readily apparent in the basal forebrain, hippocampus and cortex, and was significantly reduced by LM11A-31, with no effect on amyloid levels. These studies reveal that p75(NTR) is an important and tractable in vivo drug target for AD, with LM11A-31 representing a novel class of therapeutic candidates.
Collapse
Affiliation(s)
- Juliet K. Knowles
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA, USA
- Department of Neurology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Danielle A. Simmons
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA, USA
| | - Thuy-Vi V. Nguyen
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA, USA
| | - Lilith Vander Griend
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA, USA
| | - Youmei Xie
- Department of Neurology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Hong Zhang
- Department of Pathology and Taub Institute, Columbia University, New York, NY, USA
| | - Tao Yang
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA, USA
| | - Julia Pollak
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA, USA
| | - Timothy Chang
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA, USA
| | - Ottavio Arancio
- Department of Pathology and Taub Institute, Columbia University, New York, NY, USA
| | - Marion S. Buckwalter
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA, USA
- Palo Alto Veterans Affairs Health Care System, Palo Alto, CA, USA
| | - Stephen M. Massa
- Department of Neurology, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
- Laboratory for Computational Neurochemistry and Drug Discovery, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
- Department of Neurology, University of California–San Francisco, San Francisco, CA, USA
| | - Frank M. Longo
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA, USA
| |
Collapse
|
31
|
Nickell JR, Grinevich VP, Siripurapu KB, Smith AM, Dwoskin LP. Potential therapeutic uses of mecamylamine and its stereoisomers. Pharmacol Biochem Behav 2013; 108:28-43. [PMID: 23603417 PMCID: PMC3690754 DOI: 10.1016/j.pbb.2013.04.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 04/01/2013] [Accepted: 04/03/2013] [Indexed: 12/17/2022]
Abstract
Mecamylamine (3-methylaminoisocamphane hydrochloride) is a nicotinic parasympathetic ganglionic blocker, originally utilized as a therapeutic agent to treat hypertension. Mecamylamine administration produces several deleterious side effects at therapeutically relevant doses. As such, mecamylamine's use as an antihypertensive agent was phased out, except in severe hypertension. Mecamylamine easily traverses the blood-brain barrier to reach the central nervous system (CNS), where it acts as a nicotinic acetylcholine receptor (nAChR) antagonist, inhibiting all known nAChR subtypes. Since nAChRs play a major role in numerous physiological and pathological processes, it is not surprising that mecamylamine has been evaluated for its potential therapeutic effects in a wide variety of CNS disorders, including addiction. Importantly, mecamylamine produces its therapeutic effects on the CNS at doses 3-fold lower than those used to treat hypertension, which diminishes the probability of peripheral side effects. This review focuses on the pharmacological properties of mecamylamine, the differential effects of its stereoisomers, S(+)- and R(-)-mecamylamine, and the potential for effectiveness in treating CNS disorders, including nicotine and alcohol addiction, mood disorders, cognitive impairment and attention deficit hyperactivity disorder.
Collapse
Affiliation(s)
- Justin R Nickell
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA.
| | | | | | | | | |
Collapse
|
32
|
Prior M, Dargusch R, Ehren JL, Chiruta C, Schubert D. The neurotrophic compound J147 reverses cognitive impairment in aged Alzheimer's disease mice. ALZHEIMERS RESEARCH & THERAPY 2013; 5:25. [PMID: 23673233 PMCID: PMC3706879 DOI: 10.1186/alzrt179] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 03/01/2013] [Accepted: 03/28/2013] [Indexed: 12/12/2022]
Abstract
Introduction Despite years of research, there are no disease-modifying drugs for Alzheimer's disease (AD), a fatal, age-related neurodegenerative disorder. Screening for potential therapeutics in rodent models of AD has generally relied on testing compounds before pathology is present, thereby modeling disease prevention rather than disease modification. Furthermore, this approach to screening does not reflect the clinical presentation of AD patients which could explain the failure to translate compounds identified as beneficial in animal models to disease modifying compounds in clinical trials. Clearly a better approach to pre-clinical drug screening for AD is required. Methods To more accurately reflect the clinical setting, we used an alternative screening strategy involving the treatment of AD mice at a stage in the disease when pathology is already advanced. Aged (20-month-old) transgenic AD mice (APP/swePS1ΔE9) were fed an exceptionally potent, orally active, memory enhancing and neurotrophic molecule called J147. Cognitive behavioral assays, histology, ELISA and Western blotting were used to assay the effect of J147 on memory, amyloid metabolism and neuroprotective pathways. J147 was also investigated in a scopolamine-induced model of memory impairment in C57Bl/6J mice and compared to donepezil. Details on the pharmacology and safety of J147 are also included. Results Data presented here demonstrate that J147 has the ability to rescue cognitive deficits when administered at a late stage in the disease. The ability of J147 to improve memory in aged AD mice is correlated with its induction of the neurotrophic factors NGF (nerve growth factor) and BDNF (brain derived neurotrophic factor) as well as several BDNF-responsive proteins which are important for learning and memory. The comparison between J147 and donepezil in the scopolamine model showed that while both compounds were comparable at rescuing short term memory, J147 was superior at rescuing spatial memory and a combination of the two worked best for contextual and cued memory. Conclusion J147 is an exciting new compound that is extremely potent, safe in animal studies and orally active. J147 is a potential AD therapeutic due to its ability to provide immediate cognition benefits, and it also has the potential to halt and perhaps reverse disease progression in symptomatic animals as demonstrated in these studies.
Collapse
Affiliation(s)
- Marguerite Prior
- The Salk Institute for Biological Studies, Cellular Neurobiology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Richard Dargusch
- The Salk Institute for Biological Studies, Cellular Neurobiology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jennifer L Ehren
- The Salk Institute for Biological Studies, Cellular Neurobiology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Chandramouli Chiruta
- The Salk Institute for Biological Studies, Cellular Neurobiology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - David Schubert
- The Salk Institute for Biological Studies, Cellular Neurobiology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
33
|
Overk CR, Perez SE, Ma C, Taves MD, Soma KK, Mufson EJ. Sex steroid levels and AD-like pathology in 3xTgAD mice. J Neuroendocrinol 2013; 25:131-144. [PMID: 22889357 PMCID: PMC4065422 DOI: 10.1111/j.1365-2826.2012.02374.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 08/03/2012] [Accepted: 08/07/2012] [Indexed: 12/20/2022]
Abstract
Decreases in testosterone and 17β-oestradiol (E(2)) are associated with an increased risk for Alzheimer's disease (AD), which has been attributed to an increase in β-amyloid and tau pathological lesions. Although recent studies have used transgenic animal models to test the effects of sex steroid manipulations on AD-like pathology, almost none have systematically characterised the associations between AD lesions and sex steroid levels in the blood or brain in any mutant model. The present study evaluated age-related changes in testosterone and E(2) concentrations, as well as androgen receptor (AR) and oestrogen receptor (ER) α and β expression, in brain regions displaying AD pathology in intact male and female 3xTgAD and nontransgenic (ntg) mice. We report for the first time that circulating and brain testosterone levels significantly increase in male 3xTgAD mice with age, but without changes in AR-immunoreactive (IR) cell number in the hippocampal CA1 or medial amygdala. The age-related increase in hippocampal testosterone levels correlated positively with increases in the conformational tau isoform, Alz50. These data suggest that the over-expression of human tau up-regulate the hypothalamic-pituitary-gonadal axis in these mice. Although circulating and brain E(2) levels remained stable with age in both male and female 3xTgAD and ntg mice, ER-IR cell number in the hippocampus and medial amygdala decreased with age in female transgenic mice. Furthermore, E(2) levels were significantly higher in the hippocampus than in serum, suggesting local production of E(2). Although triple transgenic mice mimic AD-like pathology, they do not fully replicate changes in human sex steroid levels, and may not be the best model for studying the effects of sex steroids on AD lesions.
Collapse
Affiliation(s)
- Cassia R. Overk
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL USA 60612
| | - Sylvia E. Perez
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL USA 60612
| | - Chunqi Ma
- Department of Psychology, Department of Zoology, and Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC Canada V6T 1Z4
| | - Matthew D. Taves
- Department of Psychology, Department of Zoology, and Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC Canada V6T 1Z4
| | - Kiran K. Soma
- Department of Psychology, Department of Zoology, and Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC Canada V6T 1Z4
| | - Elliott J. Mufson
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL USA 60612
| |
Collapse
|
34
|
Stranahan AM, Mattson MP. Metabolic reserve as a determinant of cognitive aging. J Alzheimers Dis 2012; 30 Suppl 2:S5-13. [PMID: 22045480 DOI: 10.3233/jad-2011-110899] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mild cognitive impairment (MCI) and Alzheimer's disease (AD) represent points on a continuum of cognitive performance in aged populations. Cognition may be impaired or preserved in the context of brain aging. One theory to account for memory maintenance in the context of extensive pathology involves 'cognitive reserve', or the ability to compensate for neuropathology through greater recruitment of remaining neurons. In this review, we propose a complementary hypothesis of 'metabolic reserve', where a brain with high metabolic reserve is characterized by the presence of neuronal circuits that respond adaptively to perturbations in cellular and somatic energy metabolism and thereby protects against declining cognition. Lifestyle determinants of metabolic reserve, such as exercise, reduced caloric intake, and intake of specific dietary components can promote neuroprotection, while pathological states arising from sedentary lifestyles and excessive caloric intake contribute to neuronal endangerment. This bidirectional relationship between metabolism and cognition may be mediated by alterations in central insulin and neurotrophic factor signaling and glucose metabolism, with downstream consequences for accumulation of amyloid-β and hyperphosphorylated tau. The metabolic reserve hypothesis is supported by epidemiological findings and the spectrum of individual cognitive trajectories during aging, with additional data from animal models identifying potential mechanisms for this relationship. Identification of biomarkers for metabolic reserve could assist in generating a predictive model for the likelihood of cognitive decline with aging.
Collapse
Affiliation(s)
- Alexis M Stranahan
- Physiology Department, Georgia Health Sciences University, Augusta, Georgia, GA 30912, USA.
| | | |
Collapse
|
35
|
Müller M, Triaca V, Besusso D, Costanzi M, Horn JM, Koudelka J, Geibel M, Cestari V, Minichiello L. Loss of NGF-TrkA signaling from the CNS is not sufficient to induce cognitive impairments in young adult or intermediate-aged mice. J Neurosci 2012; 32:14885-98. [PMID: 23100411 PMCID: PMC6704821 DOI: 10.1523/jneurosci.2849-12.2012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 07/31/2012] [Accepted: 08/20/2012] [Indexed: 01/19/2023] Open
Abstract
Many molecules expressed in the CNS contribute to cognitive functions either by modulating neuronal activity or by mediating neuronal trophic support and/or connectivity. An ongoing discussion is whether signaling of nerve growth factor (NGF) through its high-affinity receptor TrkA contributes to attention behavior and/or learning and memory, based on its expression in relevant regions of the CNS such as the hippocampus, cerebral cortex, amygdala and basal forebrain. Previous animal models carrying either a null allele or transgenic manipulation of Ngf or Trka have proved difficult in addressing this question. To overcome this problem, we conditionally deleted Ngf or Trka from the CNS. Our findings confirm that NGF-TrkA signaling supports survival of only a small proportion of cholinergic neurons during development; however, this signaling is not required for trophic support or connectivity of the remaining basal forebrain cholinergic neurons. Moreover, comprehensive behavioral analysis of young adult and intermediate-aged mice lacking NGF-TrkA signaling demonstrates that this signaling is dispensable for both attention behavior and various aspects of learning and memory.
Collapse
Affiliation(s)
- Markus Müller
- Mouse Biology Unit, European Molecular Biology Laboratory, 00015 Monterotondo, Italy
| | - Viviana Triaca
- Mouse Biology Unit, European Molecular Biology Laboratory, 00015 Monterotondo, Italy
| | - Dario Besusso
- Mouse Biology Unit, European Molecular Biology Laboratory, 00015 Monterotondo, Italy
- Centre for Neuroregeneration, University of Edinburgh, EH16 4SB Edinburgh, United Kingdom
| | - Marco Costanzi
- Cellular Biology and Neurobiology Institute, Consiglio Nazionale delle Ricerche, 00143 Rome, Italy
- Department of Human Science, Lumsa University, 00193 Rome, Italy
| | - Jacqueline M. Horn
- Centre for Neuroregeneration, University of Edinburgh, EH16 4SB Edinburgh, United Kingdom
| | - Juraj Koudelka
- Centre for Neuroregeneration, University of Edinburgh, EH16 4SB Edinburgh, United Kingdom
| | - Mirjam Geibel
- Centre for Neuroregeneration, University of Edinburgh, EH16 4SB Edinburgh, United Kingdom
| | - Vincenzo Cestari
- Cellular Biology and Neurobiology Institute, Consiglio Nazionale delle Ricerche, 00143 Rome, Italy
- Department of Human Science, Lumsa University, 00193 Rome, Italy
| | - Liliana Minichiello
- Mouse Biology Unit, European Molecular Biology Laboratory, 00015 Monterotondo, Italy
- Centre for Neuroregeneration, University of Edinburgh, EH16 4SB Edinburgh, United Kingdom
| |
Collapse
|
36
|
Neuronal LR11 expression does not differentiate between clinically-defined Alzheimer's disease and control brains. PLoS One 2012; 7:e40527. [PMID: 22927900 PMCID: PMC3424248 DOI: 10.1371/journal.pone.0040527] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Accepted: 06/12/2012] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly. Because the pathological changes underlying this disease can begin decades prior to the onset of cognitive impairment, identifying the earliest events in the AD pathological cascade has critical implications for both the diagnosis and treatment of this disease. We previously reported that compared to autopsy confirmed healthy control brain, expression of LR11 (or SorLA) is markedly reduced in AD brain as well as in a subset of people with mild cognitive impairment (MCI), a prodromal clinical stage of AD. Recent studies of the LR11 gene SORL1 have suggested that the association between SORL1 single nucleotide polymorphisms (SNPs) and AD risk may not be universal. Therefore, we sought to confirm our earlier findings in a population chosen solely based on clinical criteria, as in most genetic studies. Quantitative immunohistochemistry was used to measure LR11 expression in 43 cases from the Religious Orders Study that were chosen based on a final pre-mortem clinical diagnosis of MCI, mild/moderate AD or no cognitive impairment (NCI). LR11 expression was highly variable in all three diagnostic groups, with no significant group differences. Low LR11 cases were identified using the lowest tertile of LR11 expression observed across all cases as a threshold. Contrary to previous reports, low LR11 expression was found in only 29% of AD cases. A similar proportion of both the MCI and NCI cases also displayed low LR11 expression. AD-associated lesions were present in the majority of cases regardless of diagnostic group, although we found no association between LR11 levels and pathological variables. These findings suggest that the relationship between LR11 expression and the development of AD may be more complicated than originally believed.
Collapse
|
37
|
Chu Y, Morfini GA, Langhamer LB, He Y, Brady ST, Kordower JH. Alterations in axonal transport motor proteins in sporadic and experimental Parkinson's disease. ACTA ACUST UNITED AC 2012; 135:2058-73. [PMID: 22719003 DOI: 10.1093/brain/aws133] [Citation(s) in RCA: 211] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The progressive loss of the nigrostriatal pathway is a distinguishing feature of Parkinson's disease. As terminal field loss seems to precede cell body loss, we tested whether alterations of axonal transport motor proteins would be early features in Parkinson's disease. There was a decline in axonal transport motor proteins in sporadic Parkinson's disease that preceded other well-known nigral cell-related pathology such as phenotypic downregulation of dopamine. Reductions in conventional kinesin levels precede the alterations in dopaminergic phenotypic markers (tyrosine hydroxylase) in the early stages of Parkinson's disease. This reduction was significantly greater in nigral neurons containing α-synuclein inclusions. Unlike conventional kinesin, reductions in the levels of the cytoplasmic dynein light chain Tctex type 3 subunit were only observed at late Parkinson's disease stages. Reductions in levels of conventional kinesin and cytoplasmic dynein subunits were recapitulated in a rat genetic Parkinson's disease model based on over-expression of human mutant α-synuclein (A30P). Together, our data suggest that α-synuclein aggregation is a key feature associated with reductions of axonal transport motor proteins in Parkinson's disease and support the hypothesis that dopaminergic neurodegeneration following a 'dying-back' pattern involving axonal transport disruption.
Collapse
Affiliation(s)
- Yaping Chu
- Department of Neurological Sciences, Rush University Medical Centre, 1735 West Harrison Street, Chicago, IL 60612, USA
| | | | | | | | | | | |
Collapse
|
38
|
TrkA gene ablation in basal forebrain results in dysfunction of the cholinergic circuitry. J Neurosci 2012; 32:4065-79. [PMID: 22442072 DOI: 10.1523/jneurosci.6314-11.2012] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Dysfunction of basal forebrain cholinergic neurons (BFCNs) is an early pathological hallmark of Alzheimer's disease (AD). Numerous studies have indicated that nerve growth factor (NGF) supports survival and phenotypic differentiation of BFCNs. Consistent with a potential link to AD pathogenesis, TrkA, a NGF receptor, is expressed in cholinergic forebrain neuronal populations including those in BF and striatum, and is markedly reduced in individuals with mild cognitive impairment (MCI) without dementia and early-stage AD. To investigate the role of TrkA in the development, connectivity, and function of the BF cholinergic system and its contribution to AD pathology, we have generated a forebrain-specific conditional TrkA knock-out mouse line. Our findings show a key role for TrkA signaling in establishing the BF cholinergic circuitry through the ERK pathway, and demonstrate that the normal developmental increase of choline acetyltransferase expression becomes critically dependent on TrkA signaling before neuronal connections are established. Moreover, the anatomical and physiological deficits caused by lack of TrkA signaling in BFCNs have selective impact on cognitive activity. These data demonstrate that TrkA loss results in cholinergic BF dysfunction and cognitive decline that is reminiscent of MCI and early AD.
Collapse
|
39
|
|
40
|
Lentiviral infection of rhesus macaques causes long-term injury to cortical and hippocampal projections of prostaglandin-expressing cholinergic basal forebrain neurons. J Neuropathol Exp Neurol 2012; 71:15-27. [PMID: 22157616 DOI: 10.1097/nen.0b013e31823cfac5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The simian immunodeficiency virus (SIV) macaque model resembles human immunodeficiency virus-acquired immunodeficiency syndrome (AIDS) and associated brain dysfunction. Altered expression of synaptic markers and transmitters in neuro-AIDS has been reported, but limited data exist for the cholinergic system and lipid mediators such as prostaglandins. Here, we analyzed cholinergic basal forebrain neurons with their telencephalic projections and the rate-limiting enzymes for prostaglandin synthesis, cyclooxygenase isotypes 1 and 2 (COX1 and COX2) in the brains of SIV-infected macaques with or without encephalitis and antiretroviral therapy and uninfected controls.Cyclooxygenase isotype 1, but not COX2, was coexpressed with markers of cholinergic phenotype, that is, choline acetyltransferase and vesicular acetylcholine transporter (VAChT), in basal forebrain neurons of monkey, as well as human, brain. Cyclooxygenase isotype 1 was decreased in basal forebrain neurons in macaques with AIDS versus uninfected and asymptomatic SIV-infected macaques. The VAChT-positive fiber density was reduced in frontal, parietal, and hippocampal-entorhinal cortex. Although brain SIV burden and associated COX1- and COX2-positive mononuclear and endothelial inflammatory reactions were mostly reversed in AIDS-diseased macaques that received 6-chloro-2',3'-dideoxyguanosine treatment, decreased VAChT-positive terminal density and reduced cholinergic COX1 expression were not. Thus, COX1 expression is a feature of primate cholinergic basal forebrain neurons; it may be functionally important and a critical biomarker of cholinergic dysregulation accompanying lentiviral encephalopathy. These results further imply that insufficiently prompt initiation of antiretroviral therapy in lentiviral infection may lead to neurostructurally unremarkable but neurochemically prominent irreversible brain damage.
Collapse
|
41
|
Overk CR, Lu PY, Wang YT, Choi J, Shaw JW, Thatcher GR, Mufson EJ. Effects of aromatase inhibition versus gonadectomy on hippocampal complex amyloid pathology in triple transgenic mice. Neurobiol Dis 2011; 45:479-87. [PMID: 21945538 DOI: 10.1016/j.nbd.2011.08.035] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 08/15/2011] [Accepted: 08/31/2011] [Indexed: 12/26/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia among the elderly with women exhibiting a higher risk than men for the disease. Due to these gender differences, there is great interest in the role that estrogens play in cognitive impairment and the onset of the classic amyloid and tau lesions in AD. Human and rodent studies indicate a strong association between low brain aromatase, sex hormone levels, and beta amyloid deposition. Therefore, the effects of depleting both circulating and brain estrogen levels, through gonadectomy and/or treatment with the aromatase inhibitor, anastrozole, upon hippocampal AD-like pathology in male and female 3xTgAD mice were evaluated. Liquid chromatography-mass spectrometry revealed anastrozole serum levels of 10.19 ng/mL and for the first time brain levels were detected at 4.7 pg/mL. Densitometric analysis of the hippocampus revealed that anastrozole significantly increased Aβ- but not APP/Aβ-immunoreactivity in intact 3xTgAD females compared to controls (p<0.001). Moreover, anastrozole significantly increased the number of Aβ- compared to APP/Aβ-positive hippocampal CA1 neurons in intact and gonadectomized female mice. Concurrently, anastrozole significantly reduced the APP/Aβ plaque load in 9 month old female 3xTgAD mice. These data suggest that anastrozole treatment differentially affects select amyloid species which in turn may play a role in the extraneuronal to intraneuronal deposition of this peptide.
Collapse
Affiliation(s)
- Cassia R Overk
- Department of Neurological Sciences, Rush University Medical Center Chicago, IL 60612, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Reyes JF, Fu Y, Vana L, Kanaan NM, Binder LI. Tyrosine nitration within the proline-rich region of Tau in Alzheimer's disease. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:2275-85. [PMID: 21514440 DOI: 10.1016/j.ajpath.2011.01.030] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Revised: 12/14/2010] [Accepted: 01/12/2011] [Indexed: 12/24/2022]
Abstract
A substantial body of evidence suggests that nitrative injury contributes to neurodegeneration in Alzheimer's disease (AD) and other neurodegenerative disorders. Previously, we showed in vitro that within the tau protein the N-terminal tyrosine residues (Y18 and Y29) are more susceptible to nitrative modifications than other tyrosine sites (Y197 and Y394). Using site-specific antibodies to nitrated tau at Y18 and Y29, we identified tau nitrated in both glial (Y18) and neuronal (Y29) tau pathologies. In this study, we report the characterization of two novel monoclonal antibodies, Tau-nY197 and Tau-nY394, recognizing tau nitrated at Y197 and Y394, respectively. By Western blot analysis, Tau-nY197 labeled soluble tau and insoluble paired helical filament proteins (PHF-tau) nitrated at Y197 from control and AD brain samples. Tau-nY394 failed to label soluble tau isolated from control or severe AD samples, but labeled insoluble PHF-tau to a limited extent. Immunohistochemical analysis using Tau-nY197 revealed the hallmark tau pathology associated with AD; Tau-nY394 did not detect any pathological lesions characteristic of the disorder. These data suggest that a subset of the hallmark pathological inclusions of AD contain tau nitrated at Y197. However, nitration at Y197 was also identified in soluble tau from all control samples, including those at Braak stage 0, suggesting that nitration at this site in the proline-rich region of tau may have normal biological functions in the human brain.
Collapse
Affiliation(s)
- Juan F Reyes
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | | | | | | | |
Collapse
|
43
|
Calissano P, Matrone C, Amadoro G. Apoptosis and in vitro Alzheimer disease neuronal models. Commun Integr Biol 2011; 2:163-9. [PMID: 19513272 DOI: 10.4161/cib.7704] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Accepted: 12/23/2008] [Indexed: 11/19/2022] Open
Abstract
Alzheimer disease (AD) is a human neurodegenerative disease characterized by co-existence of extracellular senile plaques (SP) and neurofibrillary tangles (NFT) associated with an extensive neuronal loss, primarily in the cerebral cortex and hippocampus. Several studies suggest that caspase(s)-mediated neuronal death occurs in cellular and animal AD models as well as in human brains of affected patients, although an etiologic role of apoptosis in such neurodegenerative disorder is still debated. This review summarizes the experimental evidences corroborating the possible involvement of apoptosis in AD pathogenesis and discusses the usefulness of ad hoc devised in vitro approaches to study how caspase(s), amyloidogenic processing and tau metabolism might reciprocally interact leading to neuronal death.
Collapse
Affiliation(s)
- P Calissano
- Institute of Neurobiology and Molecular Medicine; CNR
| | | | | |
Collapse
|
44
|
Fortress AM, Buhusi M, Helke KL, Granholm ACE. Cholinergic Degeneration and Alterations in the TrkA and p75NTR Balance as a Result of Pro-NGF Injection into Aged Rats. J Aging Res 2011; 2011:460543. [PMID: 21785728 PMCID: PMC3140182 DOI: 10.4061/2011/460543] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Accepted: 04/04/2011] [Indexed: 01/22/2023] Open
Abstract
Learning and memory impairments occurring with Alzheimer's disease (AD) are associated with degeneration of the basal forebrain cholinergic neurons (BFCNs). BFCNs extend their axons to the hippocampus where they bind nerve growth factor (NGF) which is retrogradely transported to the cell body. While NGF is necessary for BFCN survival and function via binding to the high-affinity receptor TrkA, its uncleaved precursor, pro-NGF has been proposed to induce neurodegeneration via binding to the p75NTR and its coreceptor sortilin. Basal forebrain TrkA and NGF are downregulated with aging while pro-NGF is increased. Given these data, the focus of this paper was to determine a mechanism for how pro-NGF accumulation may induce BFCN degeneration. Twenty-four hours after a single injection of pro-NGF into hippocampus, we found increased hippocampal p75NTR levels, decreased hippocampal TrkA levels, and cholinergic degeneration. The data suggest that the increase in p75NTR with AD may be mediated by elevated pro-NGF levels as a result of decreased cleavage, and that pro-NGF may be partially responsible for age-related degenerative changes observed in the basal forebrain. This paper is the first in vivo evidence that pro-NGF can affect BFCNs and may do so by regulating expression of p75NTR neurotrophin receptors.
Collapse
Affiliation(s)
- Ashley M Fortress
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | |
Collapse
|
45
|
Rojas-Castañeda JC, Vigueras-Villaseñor RM, Rojas P, Chávez-Saldaña M, Gutiérrez-Pérez O, Montes S, Ríos C. Alterations induced by chronic lead exposure on the cells of circadian pacemaker of developing rats. Int J Exp Pathol 2011; 92:243-50. [PMID: 21324006 DOI: 10.1111/j.1365-2613.2011.00761.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Lead (Pb) exposure alters the temporal organization of several physiological and behavioural processes in which the suprachiasmatic nucleus (SCN) of the hypothalamus plays a fundamental role. In this study, we evaluated the effects of chronic early Pb exposure (CePbe) on the morphology, cellular density and relative optical density (OD) in the cells of the SCN of male rats. Female Wistar rats were exposed during gestation and lactation to a Pb solution containing 320 ppm of Pb acetate through drinking water. After weaning, the pups were maintained with the same drinking water until sacrificed at 90 days of age. Pb levels in the blood, hypothalamus, hippocampus and prefrontal cortex were significantly increased in the experimental group. Chronic early Pb exposure induced a significant increase in the minor and major axes and somatic area of vasoactive intestinal polypeptide (VIP)- and vasopressin (VP)-immunoreactive neurons. The density of VIP-, VP- and glial fibrillary acidic protein (GFAP)-immunoreactive cells showed a significant decrease in the experimental group. OD analysis showed a significant increase in VIP neurons of the experimental group. The results showed that CePbe induced alterations in the cells of the SCN, as evidenced by modifications in soma morphology, cellular density and OD in circadian pacemaker cells. These findings provide a morphological and cellular basis for deficits in circadian rhythms documented in Pb-exposed animals.
Collapse
|
46
|
Moon J, Chen M, Gandhy SU, Strawderman M, Levitsky DA, Maclean KN, Strupp BJ. Perinatal choline supplementation improves cognitive functioning and emotion regulation in the Ts65Dn mouse model of Down syndrome. Behav Neurosci 2010; 124:346-61. [PMID: 20528079 DOI: 10.1037/a0019590] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In addition to mental retardation, individuals with Down syndrome (DS) also develop the neuropathological changes typical of Alzheimer's disease (AD) and the majority of these individuals exhibit dementia. The Ts65Dn mouse model of DS exhibits key features of these disorders, including early degeneration of cholinergic basal forebrain (CBF) neurons and impairments in functions dependent on the two CBF projection systems; namely, attention and explicit memory. Herein, we demonstrate that supplementing the maternal diet with excess choline during pregnancy and lactation dramatically improved attentional function of the adult trisomic offspring. Specifically, the adult offspring of choline-supplemented Ts65Dn dams performed significantly better than unsupplemented Ts65Dn mice on a series of 5 visual attention tasks, and in fact, on some tasks did not differ from the normosomic (2N) controls. A second area of dysfunction in the trisomic animals, heightened reactivity to committing an error, was partially normalized by the early choline supplementation. The 2N littermates also benefited from increased maternal choline intake on 1 attention task. These findings collectively suggest that perinatal choline supplementation might significantly lessen cognitive dysfunction in DS and reduce cognitive decline in related neurodegenerative disorders such as AD.
Collapse
Affiliation(s)
- Jisook Moon
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Neurotrophic activity of obovatol on the cultured embryonic rat neuronal cells by increase of neurotrophin release through activation of ERK pathway. Eur J Pharmacol 2010; 649:168-76. [PMID: 20868677 DOI: 10.1016/j.ejphar.2010.09.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2010] [Revised: 07/27/2010] [Accepted: 09/14/2010] [Indexed: 11/21/2022]
Abstract
Previously, we found that obovatol, a lignan compound isolated from Magnolia officinalis, has anti-cancer, anti-inflammatory, and anxiolytic effects. Recent studies showed that honokiol, magnolol, and 4-O-methylhonokiol, lignin compounds isolated from the Magnolia family have neurotrophic activity. In this study, we examined whether or not obovatol also exhibits neurite-promoting effects on rat embryonic neuronal cells. Obovatol increased neurite outgrowth in a concentration-dependent manner. Consistent with the neurite outgrowth effect, the expression of neurite differentiation markers also increased in response to obovatol. We also found that obovatol increased levels of NGF and BDNF released into the culture medium. In addition, the combination of low concentrations of obovatol (1 and 2 μM) with NGF (50 ng/ml) or with BDNF (10 ng/ml) greatly enhanced neurite outgrowth. Subsequently, we found that obovatol increased phosphorylation of ERK. However, the neurite outgrowth, and NGF and BDNF release induced by obovatol were prevented by an ERK-specific inhibitor. These results suggest that obovatol promotes neurite outgrowth due to the increased release of neurotrophic factors via activation of the ERK pathway.
Collapse
|
48
|
Chung JY, Kim MW, Bang MS, Kim M. The effect of exercise on trkA in the contralateral hemisphere of the ischemic rat brain. Brain Res 2010; 1353:187-93. [PMID: 20599809 DOI: 10.1016/j.brainres.2010.06.057] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Revised: 06/19/2010] [Accepted: 06/22/2010] [Indexed: 11/16/2022]
Abstract
Activation of the trkA pathway has been widely accepted as a crucial factor for neuronal survival during cerebral ischemia. Findings from earlier studies have suggested that exercise changed neurotrophic factors and trk family in an ischemic rat model. In this study, we investigated the question of whether or not treadmill exercise improves motor function in an ischemic rat model, and whether or not the result is associated with trkA, responding to NGF. Adult male Sprague-Dawley rats underwent surgery for permanent ischemia, followed by either 12 days of treadmill exercise or non-exercise. We found that exercise increased the level of trkA and NGF proteins in the contralateral hemisphere and improved the motor behavior index. Our data indicate that treadmill exercise ameliorates motor performance via the elevation of trkA and NGF proteins in the contralateral hemisphere after permanent brain ischemia.
Collapse
Affiliation(s)
- Jin-Young Chung
- Department of Neurology, Seoul National University Hospital, Chongno-ku, Seoul, Korea
| | | | | | | |
Collapse
|
49
|
Calissano P, Matrone C, Amadoro G. Nerve growth factor as a paradigm of neurotrophins related to Alzheimer's disease. Dev Neurobiol 2010; 70:372-83. [PMID: 20186703 DOI: 10.1002/dneu.20759] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Converging lines of evidence on the possible connection between NGF signaling and Alzheimer's diseases (AD) are unraveling new facets which could depict this neurotrophin (NTF) in a more central role. AD animal models have provided evidence that a shortage of NGF supply may induce an AD-like syndrome. In vitro experiments, moreover, are delineating a possible temporal and causal link between APP amiloydogenic processing and altered post-translational tau modifications. After NGF signaling interruption, the pivotal upstream players of the amyloid cascade (APP, beta-secretase, and active form of gamma-secretase) are up-regulated, leading to an increased production of amyloid beta peptide (Abeta) and to its intracellular aggregation in molecular species of different sizes. Contextually, the Abeta released pool generates an autocrine toxic loop in the same healthy neurons. At the same time tau protein undergoes anomalous, GSKbeta-mediated, phosphorylation at specific pathogenetic sites (Ser262 and Thr 231), caspase(s) and calpain- I- mediated truncation, detachment from microtubules with consequent cytoskeleton collapse and axonal transport impairment. All these events are inhibited when the amyloidogenic processing is reduced by beta and gamma secretase inhibitors or anti-Abeta antibodies and appear to be causally correlated to TrkA, p75CTF, Abeta, and PS1 molecular association in an Abeta-mediated fashion. In this scenario, the so-called trophic action exerted by NGF (and possibly also by other neurotrophins) in these targets neurons is actually the result of an anti-amyloidogenic activity.
Collapse
Affiliation(s)
- P Calissano
- Institute of Neurobiology and Molecular Medicine, C.N.R. Fondazione Santa Lucia, Italy.
| | | | | |
Collapse
|
50
|
Cortical M1 receptor concentration increases without a concomitant change in function in Alzheimer's disease. J Chem Neuroanat 2010; 40:63-70. [PMID: 20347961 DOI: 10.1016/j.jchemneu.2010.03.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Revised: 03/17/2010] [Accepted: 03/17/2010] [Indexed: 01/08/2023]
Abstract
Although the M(1) muscarinic receptor is a potential therapeutic target for Alzheimer's disease (AD) based on its wide spread distribution in brain and its association with learning and memory processes, whether its receptor response is altered during the onset of AD remains unclear. A novel [(35)S]GTPgammaS binding/immunocapture assay was employed to evaluated changes in M(1) receptor function in cortical tissue samples harvested from people who had no cognitive impairment (NCI), mild cognitive impairment (MCI), or AD. M(1) function was stable across clinical groups. However, [(3)H]-oxotremorine-M radioligand binding studies revealed that the concentration of M(1) cortical receptors increased significantly between the NCI and AD groups. Although M(1) receptor function did not correlate with cognitive function based upon mini-mental status examination (MMSE) or global cognitive score (GCS), functional activity was negatively correlated with the severity of neuropathology determined by Braak staging and NIA-Reagan criteria for AD. Since M(1) agonists have the potential to modify the pathologic hallmarks of AD, as well as deficits in cognitive function in animal models of this disease, the present findings provide additional support for targeting the M(1) receptor as a potential therapeutic for AD.
Collapse
|