1
|
Reitz NL, Nunes PT, Savage LM. Adolescent alcohol exposure alters age-related progression of behavioral and neurotrophic dysfunction in the TgF344-AD model in a sex-specific manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.17.603911. [PMID: 39091885 PMCID: PMC11291002 DOI: 10.1101/2024.07.17.603911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Alzheimer's Disease (AD) and heavy alcohol use are widely prevalent and lead to brain pathology. Both alcohol-related brain damage (ABRD) and AD result in cholinergic dysfunction, reductions in hippocampal neurogenesis, and the emergence of hippocampal-dependent cognitive impairments. It is still unknown how ARBD caused during a critical developmental timepoint, such as adolescence, interacts with AD-related pathologies to accelerate disease progression later in life. The current study utilized a longitudinal design to characterize behavioral and pathological changes in a transgenic rat model of AD (TgF344-AD) following adolescent intermittent ethanol (AIE) exposure. We found that AIE accelerates cognitive decline associated with AD transgenes in female rats at 6 months of age, and male AD-rats are impaired on spatial navigation by 3-months with no additional deficits due to AIE exposure. Protein levels of various AD-pathological markers were analyzed in the dorsal and ventral hippocampus of male and female rats. The data suggests that AIE-induced alterations of the tropomyosin-related kinase A receptor (TrkA) / p75 neurotrophin receptor (p75NTR) ratio creates a brain that is vulnerable to age- and AD-related pathologies, which leads to an acceleration of cognitive decline, particularly in female rats.
Collapse
|
2
|
Lane RM, Darreh-Shori T, Junge C, Li D, Yang Q, Edwards AL, Graham DL, Moore K, Mummery CJ. Onset of Alzheimer disease in apolipoprotein ɛ4 carriers is earlier in butyrylcholinesterase K variant carriers. BMC Neurol 2024; 24:116. [PMID: 38594621 PMCID: PMC11003149 DOI: 10.1186/s12883-024-03611-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 03/26/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND The authors sought to examine the impact of the K-variant of butyrylcholinesterase (BCHE-K) carrier status on age-at-diagnosis of Alzheimer disease (AD) in APOE4 carriers. METHODS Patients aged 50-74 years with cerebrospinal fluid (CSF) biomarker-confirmed AD, were recruited to clinical trial (NCT03186989 since June 14, 2017). Baseline demographics, disease characteristics, and biomarkers were evaluated in 45 patients according to BCHE-K and APOE4 allelic status in this post-hoc study. RESULTS In APOE4 carriers (N = 33), the mean age-at-diagnosis of AD in BCHE-K carriers (n = 11) was 6.4 years earlier than in BCHE-K noncarriers (n = 22, P < .001, ANOVA). In APOE4 noncarriers (N = 12) there was no observed influence of BCHE-K. APOE4 carriers with BCHE-K also exhibited slightly higher amyloid and tau accumulations compared to BCHE-K noncarriers. A predominantly amyloid, limited tau, and limbic-amnestic phenotype was exemplified by APOE4 homozygotes with BCHE-K. In the overall population, multiple regression analyses demonstrated an association of amyloid accumulation with APOE4 carrier status (P < .029), larger total brain ventricle volume (P < .021), less synaptic injury (Ng, P < .001), and less tau pathophysiology (p-tau181, P < .005). In contrast, tau pathophysiology was associated with more neuroaxonal damage (NfL, P = .002), more synaptic injury (Ng, P < .001), and higher levels of glial activation (YKL-40, P = .01). CONCLUSION These findings have implications for the genetic architecture of prognosis in early AD, not the genetics of susceptibility to AD. In patients with early AD aged less than 75 years, the mean age-at-diagnosis of AD in APOE4 carriers was reduced by over 6 years in BCHE-K carriers versus noncarriers. The functional status of glia may explain many of the effects of APOE4 and BCHE-K on the early AD phenotype. TRIAL REGISTRATION NCT03186989 since June 14, 2017.
Collapse
Affiliation(s)
- Roger M Lane
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA.
| | - Taher Darreh-Shori
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatric, Karolinska Institutet, Stockholm, Sweden
| | - Candice Junge
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| | - Dan Li
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| | - Qingqing Yang
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| | | | | | - Katrina Moore
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| | | |
Collapse
|
3
|
Criscuolo C, Chartampila E, Ginsberg SD, Scharfman HE. Dentate Gyrus Granule Cells Show Stability of BDNF Protein Expression in Mossy Fiber Axons with Age, and Resistance to Alzheimer's Disease Neuropathology in a Mouse Model. eNeuro 2024; 11:ENEURO.0192-23.2023. [PMID: 38164567 PMCID: PMC10913042 DOI: 10.1523/eneuro.0192-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 11/03/2023] [Accepted: 11/13/2023] [Indexed: 01/03/2024] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is important in the development and maintenance of neurons and their plasticity. Hippocampal BDNF has been implicated in Alzheimer's disease (AD) because hippocampal levels in AD patients and AD animal models are often downregulated, suggesting that reduced BDNF contributes to AD. However, the location where hippocampal BDNF protein is most highly expressed, the mossy fiber (MF) axons of dentate gyrus granule cells (GCs), has been understudied, and not in controlled conditions. Therefore, we evaluated MF BDNF protein in the Tg2576 mouse model of AD. Tg2576 and wild-type (WT) mice of both sexes were examined at 2-3 months of age, when amyloid-β (Aβ) is present in neurons but plaques are absent, and 11-20 months of age, after plaque accumulation. As shown previously, WT mice exhibited high levels of MF BDNF protein. Interestingly, there was no significant decline with age in either the genotype or sex. Notably, MF BDNF protein was correlated with GC ΔFosB, a transcription factor that increases after 1-2 weeks of elevated neuronal activity. We also report the novel finding that Aβ in GCs or the GC layer was minimal even at old ages. The results indicate that MF BDNF is stable in the Tg2576 mouse, and MF BDNF may remain unchanged due to increased GC neuronal activity, since BDNF expression is well known to be activity dependent. The resistance of GCs to long-term Aβ accumulation provides an opportunity to understand how to protect vulnerable neurons from increased Aβ levels and therefore has translational implications.
Collapse
Affiliation(s)
- Chiara Criscuolo
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Department of Child & Adolescent Psychiatry, NewYork University Grossman School of Medicine, New York, NY 10016
| | - Elissavet Chartampila
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Stephen D Ginsberg
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Department of Neuroscience & Physiology, NewYork University Grossman School of Medicine, New York, NY 10016
- Psychiatry, NewYork University Grossman School of Medicine, New York, NY 10016
- NYU Neuroscience Institute, NewYork University Grossman School of Medicine, New York, NY 10016
| | - Helen E Scharfman
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Department of Child & Adolescent Psychiatry, NewYork University Grossman School of Medicine, New York, NY 10016
- Department of Neuroscience & Physiology, NewYork University Grossman School of Medicine, New York, NY 10016
- NYU Neuroscience Institute, NewYork University Grossman School of Medicine, New York, NY 10016
| |
Collapse
|
4
|
Rathee S, Sen D, Pandey V, Jain SK. Advances in Understanding and Managing Alzheimer's Disease: From Pathophysiology to Innovative Therapeutic Strategies. Curr Drug Targets 2024; 25:752-774. [PMID: 39039673 DOI: 10.2174/0113894501320096240627071400] [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: 04/19/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 07/24/2024]
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative disorder characterized by the presence of amyloid-β (Aβ) plaques and tau-containing neurofibrillary tangles, leading to cognitive and physical decline. Representing the majority of dementia cases, AD poses a significant burden on healthcare systems globally, with onset typically occurring after the age of 65. While most cases are sporadic, about 10% exhibit autosomal forms associated with specific gene mutations. Neurofibrillary tangles and Aβ plaques formed by misfolded tau proteins and Aβ peptides contribute to neuronal damage and cognitive impairment. Currently, approved drugs, such as acetylcholinesterase inhibitors and N-methyl D-aspartate receptor agonists, offer only partial symptomatic relief without altering disease progression. A promising development is using lecanemab, a humanized IgG1 monoclonal antibody, as an immune therapeutic approach. Lecanemab demonstrates selectivity for polymorphic Aβ variants and binds to large soluble Aβ aggregates, providing a potential avenue for targeted treatment. This shift in understanding the role of the adaptive immune response in AD pathogenesis opens new possibilities for therapeutic interventions aiming to address the disease's intricate mechanisms. This review aims to summarize recent advancements in understanding Alzheimer's disease pathophysiology and innovative therapeutic approaches, providing valuable insights for both researchers and clinicians.
Collapse
Affiliation(s)
- Sunny Rathee
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Debasis Sen
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Vishal Pandey
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Sanjay K Jain
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| |
Collapse
|
5
|
Bañuelos C, Kittleson JR, LaNasa KH, Galiano CS, Roth SM, Perez EJ, Long JM, Roberts MT, Fong S, Rapp PR. Cognitive Aging and the Primate Basal Forebrain Revisited: Disproportionate GABAergic Vulnerability Revealed. J Neurosci 2023; 43:8425-8441. [PMID: 37798131 PMCID: PMC10711728 DOI: 10.1523/jneurosci.0456-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 09/22/2023] [Accepted: 09/23/2023] [Indexed: 10/07/2023] Open
Abstract
Basal forebrain (BF) projections to the hippocampus and cortex are anatomically positioned to influence a broad range of cognitive capacities that are known to decline in normal aging, including executive function and memory. Although a long history of research on neurocognitive aging has focused on the role of the cholinergic basal forebrain system, intermingled GABAergic cells are numerically as prominent and well positioned to regulate the activity of their cortical projection targets, including the hippocampus and prefrontal cortex. The effects of aging on noncholinergic BF neurons in primates, however, are largely unknown. In this study, we conducted quantitative morphometric analyses in brains from young adult (6 females, 2 males) and aged (11 females, 5 males) rhesus monkeys (Macaca mulatta) that displayed significant impairment on standard tests that require the prefrontal cortex and hippocampus. Cholinergic (ChAT+) and GABAergic (GAD67+) neurons were quantified through the full rostrocaudal extent of the BF. Total BF immunopositive neuron number (ChAT+ plus GAD67+) was significantly lower in aged monkeys compared with young, largely because of fewer GAD67+ cells. Additionally, GAD67+ neuron volume was greater selectively in aged monkeys without cognitive impairment compared with young monkeys. These findings indicate that the GABAergic component of the primate BF is disproportionally vulnerable to aging, implying a loss of inhibitory drive to cortical circuitry. Moreover, adaptive reorganization of the GABAergic circuitry may contribute to successful neurocognitive outcomes.SIGNIFICANCE STATEMENT A long history of research has confirmed the role of the basal forebrain in cognitive aging. The majority of that work has focused on BF cholinergic neurons that innervate the cortical mantle. Codistributed BF GABAergic populations are also well positioned to influence cognitive function, yet little is known about this prominent neuronal population in the aged brain. In this unprecedented quantitative comparison of both cholinergic and GABAergic BF neurons in young and aged rhesus macaques, we found that neuron number is significantly reduced in the aged BF compared with young, and that this reduction is disproportionately because of a loss of GABAergic neurons. Together, our findings encourage a new perspective on the functional organization of the primate BF in neurocognitive aging.
Collapse
Affiliation(s)
- Cristina Bañuelos
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland 21224
| | - Joshua R Kittleson
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland 21224
| | - Katherine H LaNasa
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland 21224
| | - Christina S Galiano
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland 21224
| | - Stephanie M Roth
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland 21224
| | - Evelyn J Perez
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland 21224
| | - Jeffrey M Long
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland 21224
| | - Mary T Roberts
- California National Primate Research Center, University of California, Davis, Davis, California 95616
| | - Sania Fong
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland 21224
- California National Primate Research Center, University of California, Davis, Davis, California 95616
| | - Peter R Rapp
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland 21224
| |
Collapse
|
6
|
Aquilani R, Cotta Ramusino M, Maestri R, Iadarola P, Boselli M, Perini G, Boschi F, Dossena M, Bellini A, Buonocore D, Doria E, Costa A, Verri M. Several dementia subtypes and mild cognitive impairment share brain reduction of neurotransmitter precursor amino acids, impaired energy metabolism, and lipid hyperoxidation. Front Aging Neurosci 2023; 15:1237469. [PMID: 37655338 PMCID: PMC10466813 DOI: 10.3389/fnagi.2023.1237469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023] Open
Abstract
Objective Dementias and mild cognitive impairment (MCI) are associated with variously combined changes in the neurotransmitter system and signaling, from neurotransmitter synthesis to synaptic binding. The study tested the hypothesis that different dementia subtypes and MCI may share similar reductions of brain availability in amino acid precursors (AAPs) of neurotransmitter synthesis and concomitant similar impairment in energy production and increase of oxidative stress, i.e., two important metabolic alterations that impact neurotransmission. Materials and methods Sixty-five demented patients (Alzheimer's disease, AD, n = 44; frontotemporal disease, FTD, n = 13; vascular disease, VaD, n = 8), 10 subjects with MCI and 15 control subjects (CTRL) were recruited for this study. Cerebrospinal fluid (CSF) and plasma levels of AAPs, energy substrates (lactate, pyruvate), and an oxidative stress marker (malondialdehyde, MDA) were measured in all participants. Results Demented patients and subjects with MCI were similar for age, anthropometric parameters, biohumoral variables, insulin resistance (HOMA index model), and CSF neuropathology markers. Compared to age-matched CTRL, both demented patients and MCI subjects showed low CSF AAP tyrosine (precursor of dopamine and catecholamines), tryptophan (precursor of serotonin), methionine (precursor of acetylcholine) limited to AD and FTD, and phenylalanine (an essential amino acid largely used for protein synthesis) (p = 0.03 to <0.0001). No significant differences were found among dementia subtypes or between each dementia subtype and MCI subjects. In addition, demented patients and MCI subjects, compared to CTRL, had similar increases in CSF and plasma levels of pyruvate (CSF: p = 0.023 to <0.0001; plasma: p < 0.002 to <0.0001) and MDA (CSF: p < 0.035 to 0.002; plasma: p < 0.0001). Only in AD patients was the CSF level of lactate higher than in CTRL (p = 0.003). Lactate/pyruvate ratios were lower in all experimental groups than in CTRL. Conclusion AD, FTD, and VaD dementia patients and MCI subjects may share similar deficits in AAPs, partly in energy substrates, and similar increases in oxidative stress. These metabolic alterations may be due to AAP overconsumption following high brain protein turnover (leading to phenylalanine reductions), altered mitochondrial structure and function, and an excess of free radical production. All these metabolic alterations may have a negative impact on synaptic plasticity and activity.
Collapse
Affiliation(s)
- Roberto Aquilani
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Matteo Cotta Ramusino
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementia, IRCCS C. Mondino Foundation, Pavia, Italy
- Dementia Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Roberto Maestri
- Department of Biomedical Engineering of the Montescano Institute, Istituti Clinici Scientifici Maugeri IRCCS, Montescano, Italy
| | - Paolo Iadarola
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Mirella Boselli
- Neurorehabilitation Unit of the Montescano Institute, Istituti Clinici Scientifici Maugeri IRCCS, Montescano, Italy
| | - Giulia Perini
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementia, IRCCS C. Mondino Foundation, Pavia, Italy
- Dementia Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Federica Boschi
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Maurizia Dossena
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Anna Bellini
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Daniela Buonocore
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Enrico Doria
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Alfredo Costa
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementia, IRCCS C. Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Manuela Verri
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| |
Collapse
|
7
|
Criscuolo C, Chartampila E, Ginsberg SD, Scharfman HE. Stability of dentate gyrus granule cell mossy fiber BDNF protein expression with age and resistance of granule cells to Alzheimer's disease neuropathology in a mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.07.539742. [PMID: 37214931 PMCID: PMC10197599 DOI: 10.1101/2023.05.07.539742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The neurotrophin brain-derived neurotrophic factor (BDNF) is important in development and maintenance of neurons and their plasticity. Hippocampal BDNF has been implicated Alzheimer's disease (AD) because hippocampal levels in AD patients and AD animal models are consistently downregulated, suggesting that reduced BDNF contributes to AD. However, the location where hippocampal BDNF protein is most highly expressed, the mossy fiber (MF) axons of dentate gyrus (DG) granule cells (GCs), has been understudied, and never in controlled in vivo conditions. We examined MF BDNF protein in the Tg2576 mouse model of AD. Tg2576 and wild type (WT) mice of both sexes were examined at 2-3 months of age, when amyloid-β (Aβ) is present in neurons but plaques are absent, and 11-20 months of age, after plaque accumulation. As shown previously, WT mice exhibited high levels of MF BDNF protein. Interestingly, there was no significant decline with age in either genotype or sex. Notably, we found a correlation between MF BDNF protein and GC ΔFosB, a transcription factor that increases after 1-2 weeks of elevated neuronal activity. Remarkably, there was relatively little evidence of Aβ in GCs or the GC layer even at old ages. Results indicate MF BDNF is stable in the Tg2576 mouse, and MF BDNF may remain unchanged due to increased GC neuronal activity, since BDNF expression is well known to be activity-dependent. The resistance of GCs to long-term Aβ accumulation provides an opportunity to understand how to protect other vulnerable neurons from increased Aβ levels and therefore has translational implications.
Collapse
Affiliation(s)
- Chiara Criscuolo
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
- Department of Child & Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Elissavet Chartampila
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Stephen D. Ginsberg
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
- Department of Neuroscience & Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, 10016, USA
- NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Helen E. Scharfman
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
- Department of Child & Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Neuroscience & Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
| |
Collapse
|
8
|
Salasova A, Monti G, Andersen OM, Nykjaer A. Finding memo: versatile interactions of the VPS10p-Domain receptors in Alzheimer’s disease. Mol Neurodegener 2022; 17:74. [PMID: 36397124 PMCID: PMC9673319 DOI: 10.1186/s13024-022-00576-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 10/17/2022] [Indexed: 11/19/2022] Open
Abstract
The family of VPS10p-Domain (D) receptors comprises five members named SorLA, Sortilin, SorCS1, SorCS2 and SorCS3. While their physiological roles remain incompletely resolved, they have been recognized for their signaling engagements and trafficking abilities, navigating a number of molecules between endosome, Golgi compartments, and the cell surface. Strikingly, recent studies connected all the VPS10p-D receptors to Alzheimer’s disease (AD) development. In addition, they have been also associated with diseases comorbid with AD such as diabetes mellitus and major depressive disorder. This systematic review elaborates on genetic, functional, and mechanistic insights into how dysfunction in VPS10p-D receptors may contribute to AD etiology, AD onset diversity, and AD comorbidities. Starting with their functions in controlling cellular trafficking of amyloid precursor protein and the metabolism of the amyloid beta peptide, we present and exemplify how these receptors, despite being structurally similar, regulate various and distinct cellular events involved in AD. This includes a plethora of signaling crosstalks that impact on neuronal survival, neuronal wiring, neuronal polarity, and synaptic plasticity. Signaling activities of the VPS10p-D receptors are especially linked, but not limited to, the regulation of neuronal fitness and apoptosis via their physical interaction with pro- and mature neurotrophins and their receptors. By compiling the functional versatility of VPS10p-D receptors and their interactions with AD-related pathways, we aim to further propel the AD research towards VPS10p-D receptor family, knowledge that may lead to new diagnostic markers and therapeutic strategies for AD patients.
Collapse
|
9
|
Tsanov M. Basal Forebrain Impairment: Understanding the Mnemonic Function of the Septal Region Translates in Therapeutic Advances. Front Neural Circuits 2022; 16:916499. [PMID: 35712645 PMCID: PMC9194835 DOI: 10.3389/fncir.2022.916499] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
The basal forebrain is one of the three major brain circuits involved in episodic memory formation together with the hippocampus and the diencephalon. The dysfunction of each of these regions is known to cause anterograde amnesia. While the hippocampal pyramidal neurons are known to encode episodic information and the diencephalic structures are known to provide idiothetic information, the contribution of the basal forebrain to memory formation has been exclusively associated with septo-hippocampal cholinergic signaling. Research data from the last decade broadened our understanding about the role of septal region in memory formation. Animal studies revealed that septal neurons process locomotor, rewarding and attentional stimuli. The integration of these signals results in a systems model for the mnemonic function of the medial septum that could guide new therapeutic strategies for basal forebrain impairment (BFI). BFI includes the disorders characterized with basal forebrain amnesia and neurodegenerative disorders that affect the basal forebrain. Here, we demonstrate how the updated model of septal mnemonic function can lead to innovative translational treatment approaches that include pharmacological, instrumental and behavioral techniques.
Collapse
Affiliation(s)
- Marian Tsanov
- UCD School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
10
|
Gonzalez S, McHugh TLM, Yang T, Syriani W, Massa SM, Longo FM, Simmons DA. Small molecule modulation of TrkB and TrkC neurotrophin receptors prevents cholinergic neuron atrophy in an Alzheimer's disease mouse model at an advanced pathological stage. Neurobiol Dis 2021; 162:105563. [PMID: 34838668 DOI: 10.1016/j.nbd.2021.105563] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/05/2021] [Accepted: 11/22/2021] [Indexed: 12/23/2022] Open
Abstract
Degeneration of basal forebrain cholinergic neurons (BFCNs) in the nucleus basalis of Meynert (NBM) and vertical diagonal band (VDB) along with their connections is a key pathological event leading to memory impairment in Alzheimer's disease (AD). Aberrant neurotrophin signaling via Trks and the p75 neurotrophin receptor (p75NTR) contributes importantly to BFCN dystrophy. While NGF/TrkA signaling has received the most attention in this regard, TrkB and TrkC signaling also provide trophic support to BFCNs and these receptors may be well located to preserve BFCN connectivity. We previously identified a small molecule TrkB/TrkC ligand, LM22B-10, that promotes cell survival and neurite outgrowth in vitro and activates TrkB/TrkC signaling in the hippocampus of aged mice when given intranasally, but shows poor oral bioavailability. An LM22B-10 derivative, PTX-BD10-2, with improved oral bioavailability has been developed and this study examined its effects on BFCN atrophy in the hAPPLond/Swe (APPL/S) AD mouse model. Oral delivery of PTX-BD10-2 was started after appreciable amyloid and cholinergic pathology was present to parallel the clinical context, as most AD patients start treatment at advanced pathological stages. PTX-BD10-2 restored cholinergic neurite integrity in the NBM and VDB, and reduced NBM neuronal atrophy in symptomatic APPL/S mice. Dystrophy of cholinergic neurites in BF target regions, including the cortex, hippocampus, and amygdala, was also reduced with treatment. Finally, PTX-BD10-2 reduced NBM tau pathology and improved the survival of cholinergic neurons derived from human induced pluripotent stem cells (iPSCs) after amyloid-β exposure. These data provide evidence that targeting TrkB and TrkC signaling with PTX-BD10-2 may be an effective disease-modifying strategy for combating cholinergic dysfunction in AD. The potential for clinical translation is further supported by the compound's reduction of AD-related degenerative processes that have progressed beyond early stages and its neuroprotective effects in human iPSC-derived cholinergic neurons.
Collapse
Affiliation(s)
- Selena Gonzalez
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Tyne L M McHugh
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Tao Yang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Wassim Syriani
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Stephen M Massa
- Department of Neurology, Laboratory for Computational Neurochemistry and Drug Discovery, Veterans Affairs Health Care System and Department of Neurology, University of California-San Francisco, San Francisco, CA 94121, United States of America
| | - Frank M Longo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Danielle A Simmons
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America.
| |
Collapse
|
11
|
Atef RM, Abdel Fattah IO, Mahmoud OM, Abdel-Rahman GM, Salem NA. Protective effects of Rosemary extract and/or Fluoxetine on Monosodium Glutamate-induced hippocampal neurotoxicity in rat. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY 2021; 62:169-177. [PMID: 34609419 PMCID: PMC8597363 DOI: 10.47162/rjme.62.1.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The use of Monosodium Glutamate (MSG) as a food flavor enhancer is increasing worldwide despite its neurotoxic effects. Fluoxetine (FLX) and Rosemary extract (RE) are known to have beneficial neuroprotective properties. Rats were divided into five groups: control group; MSG group, rats received 2 g/kg/day intraperitoneal (i.p.) injections of MSG for seven days; RE/MSG group, rats received 50 mg/kg/day of oral RE for 28 days starting prior to MSG; FLX/MSG group, rats received 10 mg/kg/day of oral FLX for 28 days beginning before MSG; and RE/FLX/MSG group, received combined treatments as mentioned above. Rats underwent the Barnes maze test, in addition to histopathological, immunohistochemical, morphometric and ultrastructural evaluations for their hippocampi. MSG increased the number of errors and escaped latency in the Barnes maze test that was significantly minimized in the three treatment groups. The MSG group exhibited pyramidal cell (PC) degeneration, shrunken glial cells and massive vascular dilatation that were improved with RE and/or FLX treatment. The number of glial fibrillary acidic protein (GFAP)-immunopositive cells were increased, and the number of PCs was decreased in the MSG group, while these values were significantly reversed with the three treatment groups with the most significant improvement at RE/FLX/MSG one. Ultrastructurally, PCs were shrunken with degenerated nuclei, dilated endoplasmic reticulum, swollen mitochondria, and vacuolations in the MSG group that were improved with RE and/or FLX. In conclusion, the combined RE and FLX treatment can ameliorate the toxic effect of MSG on rat hippocampus probably through its antioxidant and anti-inflammatory effects.
Collapse
Affiliation(s)
- Reham Mohammed Atef
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt;
| | | | | | | | | |
Collapse
|
12
|
Genetic deletion of α7 nicotinic acetylcholine receptors induces an age-dependent Alzheimer's disease-like pathology. Prog Neurobiol 2021; 206:102154. [PMID: 34453977 DOI: 10.1016/j.pneurobio.2021.102154] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/29/2021] [Accepted: 08/18/2021] [Indexed: 11/22/2022]
Abstract
The accumulation of amyloid-beta peptide (Aβ) and the failure of cholinergic transmission are key players in Alzheimer's disease (AD). However, in the healthy brain, Aβ contributes to synaptic plasticity and memory acting through α7 subtype nicotinic acetylcholine receptors (α7nAChRs). Here, we hypothesized that the α7nAChR deletion blocks Aβ physiological function and promotes a compensatory increase in Aβ levels that, in turn, triggers an AD-like pathology. To validate this hypothesis, we studied the age-dependent phenotype of α7 knock out mice. We found that α7nAChR deletion caused an impairment of hippocampal synaptic plasticity and memory at 12 months of age, paralleled by an increase of Amyloid Precursor Protein expression and Aβ levels. This was accompanied by other classical AD features such as a hyperphosphorylation of tau at residues Ser 199, Ser 396, Thr 205, a decrease of GSK-3β at Ser 9, the presence of paired helical filaments and neurofibrillary tangles, neuronal loss and an increase of GFAP-positive astrocytes. Our findings suggest that α7nAChR malfunction might precede Aβ and tau pathology, offering a different perspective to interpret the failure of anti-Aβ therapies against AD and to find novel therapeutical approaches aimed at restoring α7nAChRs-mediated Aβ function at the synapse.
Collapse
|
13
|
Shekari A, Fahnestock M. Cholinergic neurodegeneration in Alzheimer disease mouse models. HANDBOOK OF CLINICAL NEUROLOGY 2021; 182:191-209. [PMID: 34266592 DOI: 10.1016/b978-0-12-819973-2.00013-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Cholinergic signaling is critical for cognitive function. The basal forebrain is the major cholinergic output of the central nervous system. Degeneration of basal forebrain cholinergic neurons is a hallmark of Alzheimer's disease (AD). Mouse models are invaluable tools in disease research and have been used to study AD for over 25 years. However, animal models of AD vary greatly with respect to the degree of cholinergic degeneration observed. The following review will outline the most influential animal models of AD with an emphasis on the basal forebrain cholinergic system.
Collapse
Affiliation(s)
- Arman Shekari
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Margaret Fahnestock
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
14
|
Khine WWT, Voong ML, Ng TKS, Feng L, Rane GA, Kumar AP, Kua EH, Mahendran R, Mahendran R, Lee YK. Mental awareness improved mild cognitive impairment and modulated gut microbiome. Aging (Albany NY) 2020; 12:24371-24393. [PMID: 33318317 PMCID: PMC7762482 DOI: 10.18632/aging.202277] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 11/13/2020] [Indexed: 12/12/2022]
Abstract
There is ample scientific and clinical evidence of the effects of gut microbiota on the brain but no definitive evidence that the brain can affect changes in gut microbiota under the bi-directional gut-brain axis concept. As there is no pharmacotherapeutic intervention for the early stages of cognitive decline, research has focused on cognitive stimulation in reversing or slowing the impairment. Elderly patients diagnosed with mild cognitive impairment underwent a randomized-control trial of mindful awareness practice. Neuropsychological assessments, inflammatory markers, and gut microbiota profiles were tested. Here, we report that their cognitive impairment was improved and associated with changes in gut bacterial profile. A cognition-score-dependent-abundance was observed in Ruminococcus vs Recognition Trials (RT), Digit Span Backward (DSB), Semantic Fluency Span (SFS) and Memory Domain (MD); Coprococcus vs DSB, Color Trails Test 2 (CTT2) and Block Design (BD); Parabacteroides vs DSB and SFS; Fusobacterium vs DSB and CTT2; Enterobacteriaceae vs BD and SFS; Ruminococcaceae vs DSB; Phascolarctobacterium vs MD. The study showed for the first-time, alteration in the cognitive capacity leading to the corresponding changes in microbiota profiles. This strongly suggests that signals from the different segments of brain could dictate directly or indirectly the abundances of specific gut microbes.
Collapse
Affiliation(s)
- Wei Wei Thwe Khine
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore.,Functional Foods Forum, Faculty of Medicine, University of Turku, Turku 20014, Finland
| | - Miao Lian Voong
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
| | - Ted Kheng Siang Ng
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117549, Singapore
| | - Lei Feng
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117549, Singapore.,Department of Psychological Medicine, National University Hospital, Singapore 119228, Singapore
| | - Grishma Avinash Rane
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore.,Medical Sciences Cluster, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Ee Heok Kua
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117549, Singapore.,Department of Psychological Medicine, National University Hospital, Singapore 119228, Singapore
| | - Ratha Mahendran
- Department of Surgery, National University Hospital, Singapore 119228, Singapore
| | - Rathi Mahendran
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117549, Singapore.,Department of Psychological Medicine, National University Hospital, Singapore 119228, Singapore.,Duke-NUS Medical School, Singapore 169857, Singapore
| | - Yuan-Kun Lee
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore.,Department of Surgery, National University Hospital, Singapore 119228, Singapore
| |
Collapse
|
15
|
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
|
16
|
Xie Y, Seawell J, Boesch E, Allen L, Suchy A, Longo FM, Meeker RB. Small molecule modulation of the p75 neurotrophin receptor suppresses age- and genotype-associated neurodegeneration in HIV gp120 transgenic mice. Exp Neurol 2020; 335:113489. [PMID: 33007293 DOI: 10.1016/j.expneurol.2020.113489] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/07/2020] [Accepted: 09/28/2020] [Indexed: 12/21/2022]
Abstract
The persistence of HIV in the central nervous system leads to cognitive deficits in up to 50% of people living with HIV even with systemic suppression by antiretroviral treatment. The interaction of chronic inflammation with age-associated degeneration places these individuals at increased risk of accelerated aging and other neurodegenerative diseases and no treatments are available that effectively halt these processes. The adverse effects of aging and inflammation may be mediated, in part, by an increase in the expression of the p75 neurotrophin receptor (p75NTR) which shifts the balance of neurotrophin signaling toward less protective pathways. To determine if modulation of p75NTR could modify the disease process, we treated HIV gp120 transgenic mice with a small molecule ligand designed to engage p75NTR and downregulate degenerative signaling. Daily treatment with 50 mg/kg LM11A-31 for 4 months suppressed age- and genotype-dependent activation of microglia, increased microtubule associated protein-2 (MAP-2), reduced dendritic varicosities and slowed the loss of parvalbumin immunoreactive neurons in the hippocampus. An age related accumulation of microtubule associated protein Tau was identified in the hippocampus in extracellular clusters that co-expressed p75NTR suggesting a link between Tau and p75NTR. Although the significance of the relationship between p75NTR and Tau is unclear, a decrease in Tau-1 immunoreactivity as gp120 mice entered old age (>16 months) suggests that the Tau may transition to more pathological modifications; a process blocked by LM11A-31. Overall, the effects of LM11A-31 are consistent with strong neuroprotective and anti-inflammatory actions that have significant therapeutic potential.
Collapse
Affiliation(s)
- Youmie Xie
- Department of Neurology, University of North Carolina, Chapel Hill, NC 27599, United States of America
| | - Jaimie Seawell
- Department of Neurology, University of North Carolina, Chapel Hill, NC 27599, United States of America; The Edward Via College of Osteopathic Medicine, Spartanburg, SC 29303, United States of America
| | - Emily Boesch
- School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, United States of America
| | - Lauren Allen
- Department of Neurology, University of North Carolina, Chapel Hill, NC 27599, United States of America
| | - Ashley Suchy
- Department of Neurology, University of North Carolina, Chapel Hill, NC 27599, United States of America
| | - Frank M Longo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Rick B Meeker
- Department of Neurology, University of North Carolina, Chapel Hill, NC 27599, United States of America; Neurobiology Curriculum, University of North Carolina, Chapel Hill, NC 27599, United States of America.
| |
Collapse
|
17
|
Martínez-Cué C, Rueda N. Signalling Pathways Implicated in Alzheimer's Disease Neurodegeneration in Individuals with and without Down Syndrome. Int J Mol Sci 2020; 21:E6906. [PMID: 32962300 PMCID: PMC7555886 DOI: 10.3390/ijms21186906] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023] Open
Abstract
Down syndrome (DS), the most common cause of intellectual disability of genetic origin, is characterized by alterations in central nervous system morphology and function that appear from early prenatal stages. However, by the fourth decade of life, all individuals with DS develop neuropathology identical to that found in sporadic Alzheimer's disease (AD), including the development of amyloid plaques and neurofibrillary tangles due to hyperphosphorylation of tau protein, loss of neurons and synapses, reduced neurogenesis, enhanced oxidative stress, and mitochondrial dysfunction and neuroinflammation. It has been proposed that DS could be a useful model for studying the etiopathology of AD and to search for therapeutic targets. There is increasing evidence that the neuropathological events associated with AD are interrelated and that many of them not only are implicated in the onset of this pathology but are also a consequence of other alterations. Thus, a feedback mechanism exists between them. In this review, we summarize the signalling pathways implicated in each of the main neuropathological aspects of AD in individuals with and without DS as well as the interrelation of these pathways.
Collapse
Affiliation(s)
- Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, 39011 Santander, Spain;
| | | |
Collapse
|
18
|
Tabassum S, Ahmad S, Madiha S, Shahzad S, Batool Z, Sadir S, Haider S. Free L-glutamate-induced modulation in oxidative and neurochemical profile contributes to enhancement in locomotor and memory performance in male rats. Sci Rep 2020; 10:11206. [PMID: 32641780 PMCID: PMC7343824 DOI: 10.1038/s41598-020-68041-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 05/04/2020] [Indexed: 12/30/2022] Open
Abstract
Glutamate (Glu), the key excitatory neurotransmitter in the central nervous system, is considered essential for brain functioning and has a vital role in learning and memory formation. Earlier it was considered as a harmful agent but later found to be useful for many body functions. However, studies regarding the effects of free l-Glu administration on CNS function are limited. Therefore, current experiment is aimed to monitor the neurobiological effects of free l-Glu in male rats. l-Glu was orally administered to rats for 5-weeks and changes in behavioral performance were monitored. Thereafter, brain and hippocampus were collected for oxidative and neurochemical analysis. Results showed that chronic supplementation of free l-Glu enhanced locomotor performance and cognitive function of animals which may be attributed to the improved antioxidant status and cholinergic, monoaminergic and glutamatergic neurotransmission in brain and hippocampus. Current results showed that chronic supplementation of l-Glu affects the animal behaviour and brain functioning via improving the neurochemical and redox system of brain. Free l-Glu could be a useful therapeutic agent to combat neurological disturbances however this requires further targeted studies.
Collapse
Affiliation(s)
- Saiqa Tabassum
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi, 75270, Pakistan.,Department of Biosciences, Faculty of Life Science, Shaheed Zulfiqar Ali Bhutto Institute of Science and Technology (Szabist), Karachi, Pakistan
| | - Saara Ahmad
- Department of Biological and Biomedical Sciences, Aga Khan University Hospital, Karachi, Pakistan
| | - Syeda Madiha
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Sidrah Shahzad
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Zehra Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Sadia Sadir
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Saida Haider
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
19
|
Kabir MT, Uddin MS, Mamun AA, Jeandet P, Aleya L, Mansouri RA, Ashraf GM, Mathew B, Bin-Jumah MN, Abdel-Daim MM. Combination Drug Therapy for the Management of Alzheimer's Disease. Int J Mol Sci 2020; 21:E3272. [PMID: 32380758 PMCID: PMC7246721 DOI: 10.3390/ijms21093272] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/19/2020] [Accepted: 03/19/2020] [Indexed: 02/02/2023] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia worldwide. Even though the number of AD patients is rapidly growing, there is no effective treatment for this neurodegenerative disorder. At present, implementation of effective treatment approaches for AD is vital to meet clinical needs. In AD research, priorities concern the development of disease-modifying therapeutic agents to be used in the early phases of AD and the optimization of the symptomatic treatments predominantly dedicated to the more advanced AD stages. Until now, available therapeutic agents for AD treatment only provide symptomatic treatment. Since AD pathogenesis is multifactorial, use of a multimodal therapeutic intervention addressing several molecular targets of AD-related pathological processes seems to be the most practical approach to modify the course of AD progression. It has been demonstrated through numerous studies, that the clinical efficacy of combination therapy (CT) is higher than that of monotherapy. In case of AD, CT is more effective, mostly when started early, at slowing the rate of cognitive impairment. In this review, we have covered the major studies regarding CT to combat AD pathogenesis. Moreover, we have also highlighted the safety, tolerability, and efficacy of CT in the treatment of AD.
Collapse
Affiliation(s)
- Md. Tanvir Kabir
- Department of Pharmacy, BRAC University, Dhaka 1212, Bangladesh;
| | - Md. Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka 1213, Bangladesh;
- Pharmakon Neuroscience Research Network, Dhaka 1207, Bangladesh
| | - Abdullah Al Mamun
- Department of Pharmacy, Southeast University, Dhaka 1213, Bangladesh;
- Pharmakon Neuroscience Research Network, Dhaka 1207, Bangladesh
| | - Philippe Jeandet
- Research Unit, Induced Resistance and Plant Bioprotection, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, University of Reims Champagne-Ardenne, P.O. Box 1039, 51687 Reims CEDEX 2, France;
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, F-25030 Besançon, France;
| | - Rasha A. Mansouri
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad 678557, India;
| | - May N. Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia;
| | - Mohamed M. Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
20
|
Muñoz-Arenas G, Pulido G, Treviño S, Vázquez-Roque R, Flores G, Moran C, Handal-Silva A, Guevara J, Venegas B, Díaz A. Effects of metformin on recognition memory and hippocampal neuroplasticity in rats with metabolic syndrome. Synapse 2020; 74:e22153. [PMID: 32190918 DOI: 10.1002/syn.22153] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 03/06/2020] [Accepted: 03/15/2020] [Indexed: 12/18/2022]
Abstract
Metabolic syndrome (MS) is a health problem that is characterized by body fat accumulation, hypertension, dyslipidemia, and hyperglycemia; recently, it has been demonstrated that MS also damages memory processes. The first-line drug in the treatment of MS and type 2 diabetes mellitus is metformin, which is an antihyperglycemic agent. This drug has been shown to produce neuroprotection and to improve memory processes. However, the mechanism involved in this neuroprotection is unknown. A 90-day administration of metformin improved the cognitive processes of rats with MS as evaluated by the novel object recognition test, and this finding could be explained by an increase in the neuronal spine density and spine length. We also found that metformin increased the immunoreactivity of synaptophysin, sirtuin-1, AMP-activated protein kinase, and brain-derived neuronal factor, which are important plasticity markers. We conclude that metformin is an important therapeutic agent that increases neural plasticity and protects cognitive processes. The use of this drug is important in the minimization of the damage caused by MS.
Collapse
Affiliation(s)
- Guadalupe Muñoz-Arenas
- Facultad de Ciencias Quimicas, Benemerita Universidad Autonoma de Puebla, Puebla, Mexico
| | - Guadalupe Pulido
- Facultad de Ciencias Quimicas, Benemerita Universidad Autonoma de Puebla, Puebla, Mexico
| | - Samuel Treviño
- Facultad de Ciencias Quimicas, Benemerita Universidad Autonoma de Puebla, Puebla, Mexico
| | - Rubén Vázquez-Roque
- Laboratorio de Neuropsiquiatria, Instituto de Fisiologia, Benemerita Universidad Autonoma de Puebla, Puebla, Mexico
| | - Gonzalo Flores
- Laboratorio de Neuropsiquiatria, Instituto de Fisiologia, Benemerita Universidad Autonoma de Puebla, Puebla, Mexico
| | - Carolina Moran
- Laboratorio de Histologia, Instituto de Ciencias, Benemerita Universidad Autonoma de Puebla, Puebla, Mexico
| | - Anabella Handal-Silva
- Departamento de Biologia y Toxicologia de la Reproduccion, Instituto de Ciencias, Benemerita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Jorge Guevara
- Departamento de Bioquimica, Facultad de Medicina, Universidad Nacional Autonoma de México, Ciudad de Mexico, Mexico
| | - Berenice Venegas
- Facultad de Ciencias Biologicas, Benemerita Universidad Autonoma de Puebla, Puebla, Mexico
| | - Alfonso Díaz
- Facultad de Ciencias Quimicas, Benemerita Universidad Autonoma de Puebla, Puebla, Mexico
| |
Collapse
|
21
|
Zhu Q, Zhang N, Hu N, Jiang R, Lu H, Xuan A, Long D, Chen Y. Neural stem cell transplantation improves learning and memory by protecting cholinergic neurons and restoring synaptic impairment in an amyloid precursor protein/presenilin 1 transgenic mouse model of Alzheimer's disease. Mol Med Rep 2020; 21:1172-1180. [PMID: 31922229 PMCID: PMC7002968 DOI: 10.3892/mmr.2020.10918] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 09/04/2019] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent age-related neurodegenerative disorder. It is featured by the progressive accumulation of β-amyloid (Aβ) plaques and neurofibrillary tangles. This can eventually lead to a decrease of cholinergic neurons in the basal forebrain. Stem cell transplantation is an effective treatment for neurodegenerative diseases. Previous studies have revealed that different types of stem or progenitor cells can mitigate cognition impairment in different Alzheimer's disease mouse models. However, understanding the underlying mechanisms of neural stem cell (NSC) therapies for AD requires further investigation. In the present study, the effects and the underlying mechanisms of the treatment of AD by NSCs are reported. The latter were labelled with the enhanced green fluorescent protein (EGFP) prior to implantation into the bilateral hippocampus of an amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic (Tg) mouse model of AD. It was observed that the number of basal forebrain cholinergic neurons was restored and the expression of choline acetyltransferase (ChAT) protein was increased. Moreover, the levels of synaptophysin (SYP), postsynaptic density protein 95 (PSD-95) and microtubule-associated protein (MAP-2) were significantly increased in the hippocampus of NSC-treated AD mice. Notably, spatial learning and memory were both improved after transplantation of NSCs. In conclusion, the present study revealed that NSC transplantation improved learning and memory functions in an AD mouse model. This treatment allowed repairing of basal forebrain cholinergic neurons and increased the expression of the cognition-related proteins SYP, PSD-95 and MAP-2 in the hippocampus.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Nianping Zhang
- The Teaching and Research Section of Surgery, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Nan Hu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Rongrong Jiang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Huicong Lu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Aiguo Xuan
- Department of Human Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Dahong Long
- Department of Human Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Yan Chen
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| |
Collapse
|
22
|
Ginsberg SD, Malek-Ahmadi MH, Alldred MJ, Chen Y, Chen K, Chao MV, Counts SE, Mufson EJ. Brain-derived neurotrophic factor (BDNF) and TrkB hippocampal gene expression are putative predictors of neuritic plaque and neurofibrillary tangle pathology. Neurobiol Dis 2019; 132:104540. [PMID: 31349032 PMCID: PMC6834890 DOI: 10.1016/j.nbd.2019.104540] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 07/17/2019] [Accepted: 07/22/2019] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION Downregulation of brain-derived neurotrophic factor (BDNF) and its cognate neurotrophin receptor, TrkB, were observed during the progression of dementia, but whether the Alzheimer's disease (AD) pathological lesions diffuse plaques, (DPs), neuritic plaques (NPs), and neurofibrillary tangles (NFTs) are related to this alteration remains to be clarified. METHODS Negative binomial (NB) regressions were performed using gene expression data accrued from a single population of CA1 pyramidal neurons and regional hippocampal dissections obtained from participants in the Rush Religious Orders Study (RROS). RESULTS Downregulation of Bdnf is independently associated with increased entorhinal cortex NPs. Downregulation of TrkB is independently associated with increased entorhinal cortex NFTs and CA1 NPs during the progression of AD. DISCUSSION Results indicate that BDNF and TrkB dysregulation contribute to AD neuropathology, most notably hippocampal NPs and NFTs. These data suggest attenuating BDNF/TrkB signaling deficits either at the level of BDNF, TrkB, or downstream of TrkB signaling may abrogate NPs and/or NFTs.
Collapse
Affiliation(s)
- Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States of America; Department of Psychiatry, New York University Langone Medical Center, New York, NY, United States of America; Department of Neuroscience & Physiology, New York University Langone Medical Center, New York, NY, United States of America; NYU Neuroscience Institute, New York University Langone Medical Center, New York, NY, United States of America.
| | | | - Melissa J Alldred
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States of America; Department of Psychiatry, New York University Langone Medical Center, New York, NY, United States of America
| | - Yinghua Chen
- Banner Alzheimer's Institute, Phoenix, AZ, United States of America
| | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, AZ, United States of America
| | - Moses V Chao
- Department of Psychiatry, New York University Langone Medical Center, New York, NY, United States of America; NYU Neuroscience Institute, New York University Langone Medical Center, New York, NY, United States of America; Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, NY, United States of America
| | - Scott E Counts
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, United States of America; Department of Family Medicine, Michigan State University, East Lansing, MI, United States of America; Michigan Alzheimer's Disease Core Center, Ann Arbor, MI, United States of America; Hauenstein Neurosciences Center, Mercy Health Saint Mary's Hospital, Grand Rapids, MI, United States of America
| | - Elliott J Mufson
- Department of Neurobiology and Neurology, Barrow Neurological Institute, Phoenix, AZ, United States of America
| |
Collapse
|
23
|
Mirza KB, Golden CT, Nikolic K, Toumazou C. Closed-Loop Implantable Therapeutic Neuromodulation Systems Based on Neurochemical Monitoring. Front Neurosci 2019; 13:808. [PMID: 31481864 PMCID: PMC6710388 DOI: 10.3389/fnins.2019.00808] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 07/19/2019] [Indexed: 12/29/2022] Open
Abstract
Closed-loop or intelligent neuromodulation allows adjustable, personalized neuromodulation which usually incorporates the recording of a biomarker, followed by implementation of an algorithm which decides the timing (when?) and strength (how much?) of stimulation. Closed-loop neuromodulation has been shown to have greater benefits compared to open-loop neuromodulation, particularly for therapeutic applications such as pharmacoresistant epilepsy, movement disorders and potentially for psychological disorders such as depression or drug addiction. However, an important aspect of the technique is selection of an appropriate, preferably neural biomarker. Neurochemical sensing can provide high resolution biomarker monitoring for various neurological disorders as well as offer deeper insight into neurological mechanisms. The chemicals of interest being measured, could be ions such as potassium (K+), sodium (Na+), calcium (Ca2+), chloride (Cl−), hydrogen (H+) or neurotransmitters such as dopamine, serotonin and glutamate. This review focusses on the different building blocks necessary for a neurochemical, closed-loop neuromodulation system including biomarkers, sensors and data processing algorithms. Furthermore, it also highlights the merits and drawbacks of using this biomarker modality.
Collapse
Affiliation(s)
- Khalid B Mirza
- Department of Electrical and Electronic Engineering, Centre for Bio-Inspired Technology, Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Caroline T Golden
- Department of Electrical and Electronic Engineering, Centre for Bio-Inspired Technology, Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Konstantin Nikolic
- Department of Electrical and Electronic Engineering, Centre for Bio-Inspired Technology, Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Christofer Toumazou
- Department of Electrical and Electronic Engineering, Centre for Bio-Inspired Technology, Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| |
Collapse
|
24
|
Boskovic Z, Meier S, Wang Y, Milne M, Onraet T, Tedoldi A, Coulson E. Regulation of cholinergic basal forebrain development, connectivity, and function by neurotrophin receptors. Neuronal Signal 2019; 3:NS20180066. [PMID: 32269831 PMCID: PMC7104233 DOI: 10.1042/ns20180066] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/15/2019] [Accepted: 01/15/2019] [Indexed: 12/11/2022] Open
Abstract
Cholinergic basal forebrain (cBF) neurons are defined by their expression of the p75 neurotrophin receptor (p75NTR) and tropomyosin-related kinase (Trk) neurotrophin receptors in addition to cholinergic markers. It is known that the neurotrophins, particularly nerve growth factor (NGF), mediate cholinergic neuronal development and maintenance. However, the role of neurotrophin signalling in regulating adult cBF function is less clear, although in dementia, trophic signalling is reduced and p75NTR mediates neurodegeneration of cBF neurons. Here we review the current understanding of how cBF neurons are regulated by neurotrophins which activate p75NTR and TrkA, B or C to influence the critical role that these neurons play in normal cortical function, particularly higher order cognition. Specifically, we describe the current evidence that neurotrophins regulate the development of basal forebrain neurons and their role in maintaining and modifying mature basal forebrain synaptic and cortical microcircuit connectivity. Understanding the role neurotrophin signalling plays in regulating the precision of cholinergic connectivity will contribute to the understanding of normal cognitive processes and will likely provide additional ideas for designing improved therapies for the treatment of neurological disease in which cholinergic dysfunction has been demonstrated.
Collapse
Affiliation(s)
- Zoran Boskovic
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Sonja Meier
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Yunpeng Wang
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
- College of Forensic Science, Xi’an Jiaotong University, Shaanxi, China
| | - Michael R. Milne
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Tessa Onraet
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Angelo Tedoldi
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Elizabeth J. Coulson
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
25
|
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
|
26
|
Ledreux A, Håkansson K, Carlsson R, Kidane M, Columbo L, Terjestam Y, Ryan E, Tusch E, Winblad B, Daffner K, Granholm AC, Mohammed AKH. Differential Effects of Physical Exercise, Cognitive Training, and Mindfulness Practice on Serum BDNF Levels in Healthy Older Adults: A Randomized Controlled Intervention Study. J Alzheimers Dis 2019; 71:1245-1261. [PMID: 31498125 PMCID: PMC10896591 DOI: 10.3233/jad-190756] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Previous studies have indicated that an active lifestyle is associated with better brain health and a longer life, compared to a more sedentary lifestyle. These studies, both on human and animal subjects, have typically focused on a single activity, usually physical exercise, but other activities have received an increasing interest. One proposed mechanism is that physical exercise increases levels of brain-derived neurotrophic factor (BDNF) in the brain. For the first time, the long-term effects on serum BDNF levels were compared in persons who engaged in either physical exercise training, cognitive training, or mindfulness practice during 5 weeks, and compared with an active control group. Two cohorts of healthy older individuals, one from the Boston area in the US and one from the Växjö area in Sweden, participated. A total of 146 participants were randomly assigned to one of the four groups. All interventions were structurally similar, using interactive, computer-based software that directed participants to carry out specified activities for 35 minutes/day, 5 days per week for 5 weeks. Blood samples were obtained at baseline and soon after the completion of the 5-week long intervention program, and serum BDNF levels were measured using a commercially available ELISA. Only the group that underwent cognitive training increased their serum BDNF levels after 5 weeks of training (F1,74 = 4.22, p = 0.044, partial η2 = 0.054), corresponding to an average 10% increase. These results strongly suggest that cognitive training can exert beneficial effects on brain health in an older adult population.
Collapse
Affiliation(s)
- Aurélie Ledreux
- Knoebel Institute for Healthy Aging, University of Denver, 2155 E Wesley Ave, Denver, CO, USA
| | - Krister Håkansson
- Theme Aging, Karolinska University Hospital, Stockholm, Sweden
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| | - Roger Carlsson
- Department of Psychology, Linnaeus University, Växjö, Sweden
| | - Mhretab Kidane
- Department of Computer Science and Media Technology, Linnaeus University, Växjö, Sweden
| | - Laura Columbo
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Ave, Charleston, SC, USA
| | | | - Eliza Ryan
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Erich Tusch
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| | - Kirk Daffner
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ann-Charlotte Granholm
- Knoebel Institute for Healthy Aging, University of Denver, 2155 E Wesley Ave, Denver, CO, USA
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Ave, Charleston, SC, USA
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| | - Abdul Kadir H Mohammed
- Department of Psychology, Linnaeus University, Växjö, Sweden
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
27
|
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
|
28
|
Loera-Valencia R, Piras A, Ismail MAM, Manchanda S, Eyjolfsdottir H, Saido TC, Johansson J, Eriksdotter M, Winblad B, Nilsson P. Targeting Alzheimer's disease with gene and cell therapies. J Intern Med 2018; 284:2-36. [PMID: 29582495 DOI: 10.1111/joim.12759] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) causes dementia in both young and old people affecting more than 40 million people worldwide. The two neuropathological hallmarks of the disease, amyloid beta (Aβ) plaques and neurofibrillary tangles consisting of protein tau are considered the major contributors to the disease. However, a more complete picture reveals significant neurodegeneration and decreased cell survival, neuroinflammation, changes in protein and energy homeostasis and alterations in lipid and cholesterol metabolism. In addition, gene and cell therapies for severe neurodegenerative disorders have recently improved technically in terms of safety and efficiency and have translated to the clinic showing encouraging results. Here, we review broadly current data within the field for potential targets that could modify AD through gene and cell therapy strategies. We envision that not only Aβ will be targeted in a disease-modifying treatment strategy but rather that a combination of treatments, possibly at different intervention times may prove beneficial in curing this devastating disease. These include decreased tau pathology, neuronal growth factors to support neurons and modulation of neuroinflammation for an appropriate immune response. Furthermore, cell based therapies may represent potential strategies in the future.
Collapse
Affiliation(s)
- R Loera-Valencia
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - A Piras
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - M A M Ismail
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden.,Theme Neuro, Diseases of the Nervous System Patient Flow, Karolinska University Hospital, Huddinge, Sweden
| | - S Manchanda
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - H Eyjolfsdottir
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - T C Saido
- RIKEN Brain Science Institute, Wako, Saitama, Japan
| | - J Johansson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - M Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - B Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - P Nilsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
29
|
Neurotrophins and cholinergic enzyme regulated by calpain-2: New insights into neuronal apoptosis induced by polybrominated diphenyl ether-153. Toxicol Lett 2018; 291:29-38. [DOI: 10.1016/j.toxlet.2018.03.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 02/28/2018] [Accepted: 03/31/2018] [Indexed: 02/06/2023]
|
30
|
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
|
31
|
Rajmohan R, Reddy PH. Amyloid-Beta and Phosphorylated Tau Accumulations Cause Abnormalities at Synapses of Alzheimer's disease Neurons. J Alzheimers Dis 2018; 57:975-999. [PMID: 27567878 DOI: 10.3233/jad-160612] [Citation(s) in RCA: 297] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Amyloid-beta (Aβ) and hyperphosphorylated tau are hallmark lesions of Alzheimer's disease (AD). However, the loss of synapses and dysfunctions of neurotransmission are more directly tied to disease severity. The role of these lesions in the pathoetiological progression of the disease remains contested. Biochemical, cellular, molecular, and pathological studies provided several lines of evidence and improved our understanding of how Aβ and hyperphosphorylated tau accumulation may directly harm synapses and alter neurotransmission. In vitro evidence suggests that Aβ and hyperphosphorylated tau have both direct and indirect cytotoxic effects that affect neurotransmission, axonal transport, signaling cascades, organelle function, and immune response in ways that lead to synaptic loss and dysfunctions in neurotransmitter release. Observations in preclinical models and autopsy studies support these findings, suggesting that while the pathoetiology of positive lesions remains elusive, their removal may reduce disease severity and progression. The purpose of this article is to highlight the need for further investigation of the role of tau in disease progression and its interactions with Aβ and neurotransmitters alike.
Collapse
Affiliation(s)
- Ravi Rajmohan
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Speech, Language and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
32
|
Ginsberg SD, Malek-Ahmadi MH, Alldred MJ, Che S, Elarova I, Chen Y, Jeanneteau F, Kranz TM, Chao MV, Counts SE, Mufson EJ. Selective decline of neurotrophin and neurotrophin receptor genes within CA1 pyramidal neurons and hippocampus proper: Correlation with cognitive performance and neuropathology in mild cognitive impairment and Alzheimer's disease. Hippocampus 2017; 29:422-439. [PMID: 28888073 DOI: 10.1002/hipo.22802] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 09/01/2017] [Accepted: 09/05/2017] [Indexed: 01/02/2023]
Abstract
Hippocampal CA1 pyramidal neurons, a major component of the medial temporal lobe memory circuit, are selectively vulnerable during the progression of Alzheimer's disease (AD). The cellular mechanism(s) underlying degeneration of these neurons and the relationship to cognitive performance remains largely undefined. Here, we profiled neurotrophin and neurotrophin receptor gene expression within microdissected CA1 neurons along with regional hippocampal dissections from subjects who died with a clinical diagnosis of no cognitive impairment (NCI), mild cognitive impairment (MCI), or AD using laser capture microdissection (LCM), custom-designed microarray analysis, and qPCR of CA1 subregional dissections. Gene expression levels were correlated with cognitive test scores and AD neuropathology criteria. We found a significant downregulation of several neurotrophin genes (e.g., Gdnf, Ngfb, and Ntf4) in CA1 pyramidal neurons in MCI compared to NCI and AD subjects. In addition, the neurotrophin receptor transcripts TrkB and TrkC were decreased in MCI and AD compared to NCI. Regional hippocampal dissections also revealed select neurotrophic gene dysfunction providing evidence for vulnerability within the hippocampus proper during the progression of dementia. Downregulation of several neurotrophins of the NGF family and cognate neurotrophin receptor (TrkA, TrkB, and TrkC) genes correlated with antemortem cognitive measures including the Mini-Mental State Exam (MMSE), a composite global cognitive score (GCS), and Episodic, Semantic, and Working Memory, Perceptual Speed, and Visuospatial domains. Significant correlations were found between select neurotrophic expression downregulation and neuritic plaques (NPs) and neurofibrillary tangles (NFTs), but not diffuse plaques (DPs). These data suggest that dysfunction of neurotrophin signaling complexes have profound negative sequelae within vulnerable hippocampal cell types, which play a role in mnemonic and executive dysfunction during the progression of AD.
Collapse
Affiliation(s)
- Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York.,Department of Psychiatry, New York University Langone Medical Center, New York, New York.,Department of Neuroscience & Physiology, New York University Langone Medical Center, New York, New York.,Neuroscience Institute, New York University Langone Medical Center, New York, New York
| | | | - Melissa J Alldred
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York.,Department of Psychiatry, New York University Langone Medical Center, New York, New York
| | - Shaoli Che
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York.,Department of Psychiatry, New York University Langone Medical Center, New York, New York
| | - Irina Elarova
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York
| | | | - Freddy Jeanneteau
- Inserm, U1191, Institute of Functional Genomics, Montpellier, F-34000, France.,CNRS, UMR-5203, Montpellier, F-34000, France.,Université de Montpellier, Montpellier, F-34000, France
| | - Thorsten M Kranz
- Department of Psychiatry, New York University Langone Medical Center, New York, New York.,Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, New York
| | - Moses V Chao
- Department of Psychiatry, New York University Langone Medical Center, New York, New York.,Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, New York
| | - Scott E Counts
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan.,Department of Family Medicine, Michigan State University, East Lansing, Michigan.,Michigan Alzheimer's Disease Core Center, Ann Arbor, Michigan.,Mercy Health Saint Mary's Hospital, Hauenstein Neurosciences Center, Grand Rapids, Michigan
| | - Elliott J Mufson
- Department of Neurobiology and Neurology, Barrow Neurological Institute, Phoenix, Arizona
| |
Collapse
|
33
|
Differential deregulation of NGF and BDNF neurotrophins in a transgenic rat model of Alzheimer's disease. Neurobiol Dis 2017; 108:307-323. [PMID: 28865749 DOI: 10.1016/j.nbd.2017.08.019] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 08/08/2017] [Accepted: 08/29/2017] [Indexed: 12/17/2022] Open
Abstract
Evidence from human neuropathological studies indicates that the levels of the neurotrophins nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) are compromised in Alzheimer's disease. However, the causes and temporal (pathology-dependent) evolution of these alterations are not completely understood. To elucidate these issues, we investigated the McGill-R-Thy1-APP transgenic rat, which exhibits progressive intracellular and extracellular amyloid-beta (Aβ) pathology and ensuing cognitive deficits. Neurochemical analyses revealed a differential dysregulation of NGF and BDNF transcripts and protein expression. While BDNF mRNA levels were significantly reduced at very early stages of amyloid pathology, before plaques appeared, there were no changes in NGF mRNA expression even at advanced stages. Paradoxically, the protein levels of the NGF precursor were increased. These changes in neurotrophin expression are identical to those seen during the progression of Alzheimer's disease. At advanced pathological stages, deficits in the protease cascade controlling the maturation and degradation of NGF were evident in McGill transgenic rats, in line with the paradoxical upregulation of proNGF, as seen in Alzheimer's disease, in the absence of changes in NGF mRNA. The compromise in NGF metabolism and BDNF levels was accompanied by downregulation of cortical cholinergic synapses; strengthening the evidence that neurotrophin dysregulation affects cholinergic synapses and synaptic plasticity. Our findings suggest a differential temporal deregulation of NGF and BDNF neurotrophins, whereby deficits in BDNF mRNA appear at early stages of intraneuronal Aβ pathology, before alterations in NGF metabolism and cholinergic synapse loss manifest.
Collapse
|
34
|
Pepeu G, Grazia Giovannini M. The fate of the brain cholinergic neurons in neurodegenerative diseases. Brain Res 2017; 1670:173-184. [PMID: 28652219 DOI: 10.1016/j.brainres.2017.06.023] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 06/21/2017] [Accepted: 06/22/2017] [Indexed: 01/03/2023]
Abstract
The aims of this review are: 1) to describe which cholinergic neurons are affected in brain neurodegenerative diseases leading to dementia; 2) to discuss the possible causes of the degeneration of the cholinergic neurons, 3) to summarize the functional consequences of the cholinergic deficit. The brain cholinergic system is basically constituted by three populations of phenotypically similar neurons forming a series of basal forebrain nuclei, the midpontine nuclei and a large population of striatal interneurons. In Alzheimer's disease there is an extensive loss of forebrain cholinergic neurons accompanied by a reduction of the cholinergic fiber network of the cortical mantel and hippocampus. The midpontine cholinergic nuclei are spared. The same situation occurs in the corticobasal syndrome and dementia following alcohol abuse and traumatic brain injury. Conversely, in Parkinson's disease, the midpontine nuclei degenerate, together with the dopaminergic nuclei, reducing the cholinergic input to thalamus and forebrain whereas the forebrain cholinergic neurons are spared. In Parkinson's disease with dementia, Lewis Body Dementia and Parkinsonian syndromes both groups of forebrain and midpontine cholinergic nuclei degenerate. In Huntington's disease a dysfunction of the striatal cholinergic interneurons without cell loss takes place. The formation and accumulation of misfolded proteins such as β-amyloid oligomers and plaques, tau protein tangles and α-synuclein clumps, and aggregated mutated huntingtin play a crucial role in the neuronal degeneration by direct cellular toxicity of the misfolded proteins and through the toxic compounds resulting from an extensive inflammatory reaction. Evidences indicate that β-amyloid disrupts NGF metabolism causing the degeneration of the cholinergic neurons which depend on NGF for their survival, namely the forebrain cholinergic neurons, sparing the midpontine and striatal neurons which express no specific NGF receptors. It is feasible that the latter cholinergic neurons may be damaged by direct toxicity of tau, α-synuclein and inflammations products through mechanisms not fully understood. Attention and learning and memory impairment are the functional consequences of the forebrain cholinergic neuron dysfunction, whereas the loss of midpontine cholinergic neurons results primarily in motor and sleep disturbances.
Collapse
Affiliation(s)
- Giancarlo Pepeu
- Department of Health Sciences, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy.
| | - Maria Grazia Giovannini
- Department of Health Sciences, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy.
| |
Collapse
|
35
|
Bennys K, Gabelle A, Berr C, De Verbizier D, Andrieu S, Vellas B, Touchon J. Cognitive Event-Related Potential, an Early Diagnosis Biomarker in Frail Elderly Subjects: The ERP-MAPT-PLUS Ancillary Study. J Alzheimers Dis 2017; 58:87-97. [DOI: 10.3233/jad-161012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Karim Bennys
- Memory Research and Resource Center for Alzheimer’s Disease, Department of Neurology, University Hospital of Montpellier, Montpellier, France
- Clinical Neurophysiology Unit, Department of Neurology, University Hospital of Montpellier, France
| | - Audrey Gabelle
- Memory Research and Resource Center for Alzheimer’s Disease, Department of Neurology, University Hospital of Montpellier, Montpellier, France
- INSERM U1183, University of Montpellier, France
| | | | - Delphine De Verbizier
- Department of Nuclear Medicine, Gui de Chauliac University Hospital, Montpellier, France
| | - Sandrine Andrieu
- Gérontopôle, Department of Geriatrics, CHU Toulouse, Purpan University Hospital, Toulouse, France
- INSERM UMR 1027, Toulouse, France
- University of Toulouse III, Toulouse, France
- Department of Epidemiology and Public Health, CHU Toulouse, Toulouse, France
| | - Bruno Vellas
- Gérontopôle, Department of Geriatrics, CHU Toulouse, Purpan University Hospital, Toulouse, France
- INSERM UMR 1027, Toulouse, France
- University of Toulouse III, Toulouse, France
| | - Jacques Touchon
- Memory Research and Resource Center for Alzheimer’s Disease, Department of Neurology, University Hospital of Montpellier, Montpellier, France
- INSERM U1061, University of Montpellier, France
| | | |
Collapse
|
36
|
Kwon H, Jung IH, Yi JH, Kim JH, Park JH, Lee S, Jung JW, Lee YC, Ryu JH, Kim DH. The Seed of Zizyphus jujuba var. spinosa Attenuates Alzheimer’s Disease-Associated Hippocampal Synaptic Deficits through BDNF/TrkB Signaling. Biol Pharm Bull 2017; 40:2096-2104. [DOI: 10.1248/bpb.b17-00378] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Huiyoung Kwon
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University
| | - In Ho Jung
- Department of Life and Nanopharmaceutical Science, Kyung Hee University
| | - Ji Hyun Yi
- School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol
| | - Jae Hoon Kim
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University
| | - Jung Hoon Park
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University
| | - Seungheon Lee
- Department of Aquatic Biomedical Sciences, School of Marine Biomedical Science, College of Ocean Science, Jeju National University
| | - Ji Wook Jung
- Department of Herbal Medicinal Pharmacology, College of Herbal Bio-Industry, Daegu Haany University
| | - Young Choon Lee
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University
- Institute of Convergence Bio-Health, Dong-A University
| | - Jong Hoon Ryu
- Department of Life and Nanopharmaceutical Science, Kyung Hee University
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University
- Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University
| | - Dong Hyun Kim
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University
- Institute of Convergence Bio-Health, Dong-A University
| |
Collapse
|
37
|
Kelley CM, Ash JA, Powers BE, Velazquez R, Alldred MJ, Ikonomovic MD, Ginsberg SD, Strupp BJ, Mufson EJ. Effects of Maternal Choline Supplementation on the Septohippocampal Cholinergic System in the Ts65Dn Mouse Model of Down Syndrome. Curr Alzheimer Res 2016; 13:84-96. [PMID: 26391045 DOI: 10.2174/1567205012666150921100515] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 09/01/2015] [Accepted: 09/10/2015] [Indexed: 01/07/2023]
Abstract
Down syndrome (DS), caused by trisomy of chromosome 21, is marked by intellectual disability (ID) and early onset of Alzheimer's disease (AD) neuropathology including hippocampal cholinergic projection system degeneration. Here we determined the effects of age and maternal choline supplementation (MCS) on hippocampal cholinergic deficits in Ts65Dn mice compared to 2N mice sacrificed at 6-8 and 14-18 months of age. Ts65Dn mice and disomic (2N) littermates sacrificed at ages 6-8 and 14-18 mos were used for an aging study and Ts65Dn and 2N mice derived from Ts65Dn dams were maintained on either a choline-supplemented or a choline-controlled diet (conception to weaning) and examined at 14-18 mos for MCS studies. In the latter, mice were behaviorally tested on the radial arm Morris water maze (RAWM) and hippocampal tissue was examined for intensity of choline acetyltransferase (ChAT) immunoreactivity. Hippocampal ChAT activity was evaluated in a separate cohort. ChAT-positive fiber innervation was significantly higher in the hippocampus and dentate gyrus in Ts65Dn mice compared with 2N mice, independent of age or maternal diet. Similarly, hippocampal ChAT activity was significantly elevated in Ts65Dn mice compared to 2N mice, independent of maternal diet. A significant increase with age was seen in hippocampal cholinergic innervation of 2N mice, but not Ts65Dn mice. Degree of ChAT intensity correlated negatively with spatial memory ability in unsupplemented 2N and Ts65Dn mice, but positively in MCS 2N mice. The increased innervation produced by MCS appears to improve hippocampal function, making this a therapy that may be exploited for future translational approaches in human DS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Elliott J Mufson
- Barrow Neurological Institute, Dept. Neurobiology, Phoenix, AZ 85031, USA.
| |
Collapse
|
38
|
Deibel S, Weishaupt N, Regis A, Hong N, Keeley R, Balog R, Bye C, Himmler S, Whitehead S, McDonald R. Subtle learning and memory impairment in an idiopathic rat model of Alzheimer's disease utilizing cholinergic depletions and β-amyloid. Brain Res 2016; 1646:12-24. [DOI: 10.1016/j.brainres.2016.05.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 02/02/2023]
|
39
|
Current pharmacotherapy and putative disease-modifying therapy for Alzheimer's disease. Neurol Sci 2016; 37:1403-35. [PMID: 27250365 DOI: 10.1007/s10072-016-2625-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/24/2016] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease of the central nervous system correlated with the progressive loss of cognition and memory. β-Amyloid plaques, neurofibrillary tangles and the deficiency in cholinergic neurotransmission constitute the major hallmarks of the AD. Two major hypotheses have been implicated in the pathogenesis of AD namely the cholinergic hypothesis which ascribed the clinical features of dementia to the deficit cholinergic neurotransmission and the amyloid cascade hypothesis which emphasized on the deposition of insoluble peptides formed due to the faulty cleavage of the amyloid precursor protein. Current pharmacotherapy includes mainly the acetylcholinesterase inhibitors and N-methyl-D-aspartate receptor agonist which offer symptomatic therapy and does not address the underlying cause of the disease. The disease-modifying therapy has garnered a lot of research interest for the development of effective pharmacotherapy for AD. β and γ-Secretase constitute attractive targets that are focussed in the disease-modifying approach. Potentiation of α-secretase also seems to be a promising approach towards the development of an effective anti-Alzheimer therapy. Additionally, the ameliorative agents that prevent aggregation of amyloid peptide and also the ones that modulate inflammation and oxidative damage associated with the disease are focussed upon. Development in the area of the vaccines is in progress to combat the characteristic hallmarks of the disease. Use of cholesterol-lowering agents also is a fruitful strategy for the alleviation of the disease as a close association between the cholesterol and AD has been cited. The present review underlines the major therapeutic strategies for AD with focus on the new developments that are on their way to amend the current therapeutic scenario of the disease.
Collapse
|
40
|
Grothe MJ, Heinsen H, Amaro E, Grinberg LT, Teipel SJ. Cognitive Correlates of Basal Forebrain Atrophy and Associated Cortical Hypometabolism in Mild Cognitive Impairment. Cereb Cortex 2016; 26:2411-2426. [PMID: 25840425 PMCID: PMC4869802 DOI: 10.1093/cercor/bhv062] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Degeneration of basal forebrain (BF) cholinergic nuclei is associated with cognitive decline, and this effect is believed to be mediated by neuronal dysfunction in the denervated cortical areas. MRI-based measurements of BF atrophy are increasingly being used as in vivo surrogate markers for cholinergic degeneration, but the functional implications of reductions in BF volume are not well understood. We used high-resolution MRI, fluorodeoxyglucose-positron emission tomography (PET), and neuropsychological test data of 132 subjects with mild cognitive impairment (MCI) and 177 cognitively normal controls to determine associations between BF atrophy, cortical hypometabolism, and cognitive deficits. BF atrophy in MCI correlated with both impaired memory function and attentional control deficits, whereas hippocampus volume was more specifically associated with memory deficits. BF atrophy was also associated with widespread cortical hypometabolism, and path analytic models indicated that hypometabolism in domain-specific cortical networks mediated the association between BF volume and cognitive dysfunction. The presence of cortical amyloid pathology, as assessed using AV45-PET, did not significantly interact with the observed associations. These data underline the potential of multimodal imaging markers to study structure-function-cognition relationships in the living human brain and provide important in vivo evidence for an involvement of the human BF in cortical activity and cognitive function.
Collapse
Affiliation(s)
- Michel J. Grothe
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
| | - Helmut Heinsen
- Laboratory of Morphological Brain Research, Department of Psychiatry, University of Würzburg, Würzburg, Germany
| | | | - Lea T. Grinberg
- Aging Brain Study Group, LIM-22, Department of Pathology, University of Sao Paulo Medical School, Sao Paulo, Brazil
- UCSF Memory and Aging Center, University of California – San Francisco, San Francisco, CA, USA
| | - Stefan J. Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| |
Collapse
|
41
|
Madhavadas S, Kapgal VK, Kutty BM, Subramanian S. The Neuroprotective Effect of Dark Chocolate in Monosodium Glutamate-Induced Nontransgenic Alzheimer Disease Model Rats: Biochemical, Behavioral, and Histological Studies. J Diet Suppl 2015; 13:449-60. [PMID: 26673833 DOI: 10.3109/19390211.2015.1108946] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The vulnerability to oxidative stress and cognitive decline continue to increase during both normal and pathological aging. Dietary changes and sedentary life style resulting in mid-life obesity and type 2 diabetes, if left uncorrected, further add to the risk of cognitive decline and Alzheimer disease (AD) in the later stages of life. Certain antioxidant agents such as dietary polyphenols, taken in adequate quantities, have been suggested to improve the cognitive processes. In this study, we examined the effect of oral administration of dark chocolate (DC) containing 70% cocoa solids and 4% total polyphenol content for three months at a dose of 500 mg/Kg body weight per day to 17-month-old monosodium glutamate treated obese Sprague-Dawley rats, earlier characterized as a nontransgenic AD (NTAD) rat model after reversal of obesity, diabetes, and consequent cognitive impairments. The results demonstrated that DC reduced the hyperglycemia, inhibited the cholinesterase activity in the hippocampal tissue homogenates, and improved the cognitive performance in spatial memory related Barnes maze task. Histological studies revealed an increase in cell volume in the DC treated rats in the CA3 region of the hippocampus. These findings demonstrated the benefits of DC in enhancing cognitive function and cholinergic activity in the hippocampus of the aged NTAD rats while correcting their metabolic disturbances.
Collapse
Affiliation(s)
- Sowmya Madhavadas
- a Department of Neurochemistry, National Institute of Mental Health & Neurosciences , Bangalore , India
| | - Vijaya Kumar Kapgal
- b Department of Neurophysiology, National Institute of Mental Health & Neurosciences , Bangalore , India
| | - Bindu M Kutty
- b Department of Neurophysiology, National Institute of Mental Health & Neurosciences , Bangalore , India
| | - Sarada Subramanian
- a Department of Neurochemistry, National Institute of Mental Health & Neurosciences , Bangalore , India
| |
Collapse
|
42
|
Williams KS, Killebrew DA, Clary GP, Meeker RB. Opposing Effects of NGF and proNGF on HIV Induced Macrophage Activation. J Neuroimmune Pharmacol 2015; 11:98-120. [PMID: 26420421 DOI: 10.1007/s11481-015-9631-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/01/2015] [Indexed: 02/04/2023]
Abstract
Macrophage and microglial activation by HIV in the central nervous system (CNS) triggers the secretion of soluble factors which damage neurons. Therapeutic approaches designed to restore cognitive function by suppressing this inflammatory activity have not yet been successful. Recent studies have indicated that the phenotype of macrophages is differentially controlled by the mature and pro form of nerve growth factor. These cells therefore may be highly responsive to the imbalance in pro versus mature neurotrophins often associated with neurodegenerative diseases. In this study we evaluated the interactions between neurotrophins and HIV induced macrophage activation. HIV stimulation of macrophages induced a neurotoxic phenotype characterized by the expression of podosomes, suppression of calcium spiking and increased neurotoxin production. The secretome of the activated macrophages revealed a bias toward anti-angiogenic like activity and increased secretion of MMP-9. Co-stimulation with NGF and HIV suppressed neurotoxin secretion, increased calcium spiking, suppressed podosome expression and reversed 86% of the proteins secreted in response to HIV, including MMP-9 and many growth factors. In contrast, co-stimulation of macrophages with proNGF not only failed to reverse the effects of HIV but increased the neurotoxic phenotype. These differential effects of proNGF and NGF on HIV activation provide a potential novel therapeutic avenue for controlling macrophage activation in response to HIV.
Collapse
Affiliation(s)
- Kimberly S Williams
- Department of Neurology, CB #7025, University of North Carolina, 6109F Neuroscience Research Building, 115 Mason Farm Road, Chapel Hill, NC, 27599, USA
- Neurobiology Curriculum, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Deirdre A Killebrew
- Department of Neurology, CB #7025, University of North Carolina, 6109F Neuroscience Research Building, 115 Mason Farm Road, Chapel Hill, NC, 27599, USA
- Center for Science and Mathematics Education, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Gillian P Clary
- Department of Neurology, CB #7025, University of North Carolina, 6109F Neuroscience Research Building, 115 Mason Farm Road, Chapel Hill, NC, 27599, USA
- US Food and Drug Administration, Rockville, MD, 20852, USA
| | - Rick B Meeker
- Department of Neurology, CB #7025, University of North Carolina, 6109F Neuroscience Research Building, 115 Mason Farm Road, Chapel Hill, NC, 27599, USA.
- Neurobiology Curriculum, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
43
|
Alvarez A, Aleixandre M, Linares C, Masliah E, Moessler H. Apathy and APOE4 are associated with reduced BDNF levels in Alzheimer's disease. J Alzheimers Dis 2015; 42:1347-55. [PMID: 25024337 DOI: 10.3233/jad-140849] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Reduced brain-derived neurotrophic factor (BDNF) signaling is considered as a pathogenic event in early Alzheimer's disease (AD), but the influence of apathy and apolipoprotein E ε4 allele (APOE4) on serum BDNF values was not previously investigated in AD. We evaluated serum BDNF levels in AD, amnestic mild cognitive impairment (MCI), and control subjects. Baseline BDNF levels were similar in AD, MCI, and controls. AD patients having apathy showed lower BDNF values than patients without apathy (p < 0.05). After correction for the influence of apathy, APOE4 carriers showed lower BDNF levels (p < 0.01) and MMSE scores (p < 0.01) than non-APOE4 carriers in the subgroup of AD females, but not in males. Significant (p < 0.05) positive correlations between BDNF values and MMSE scores were only observed in subgroups of AD males and of AD patients without apathy. These results are showing the association of apathy and APOE4 with reduced serum BDNF levels in AD, and are suggesting that BDNF reductions might contribute to the worse cognitive performance exhibited by AD apathetic patients and female APOE4 carriers.
Collapse
Affiliation(s)
- Antón Alvarez
- Medinova Institute of Neurosciences, Clínica RehaSalud, A Coruña, Spain
| | | | | | - Eliezer Masliah
- Departments of Neurosciences and Pathology, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | | |
Collapse
|
44
|
Keeley R, Hong N, Fisher A, McDonald R. Co-morbid beta-amyloid toxicity and stroke produce impairments in an ambiguous context task in rats without any impairment in spatial working memory. Neurobiol Learn Mem 2015; 119:42-51. [DOI: 10.1016/j.nlm.2015.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 01/02/2015] [Accepted: 01/05/2015] [Indexed: 01/28/2023]
|
45
|
Bernard-Gauthier V, Aliaga A, Aliaga A, Boudjemeline M, Hopewell R, Kostikov A, Rosa-Neto P, Thiel A, Schirrmacher R. Syntheses and evaluation of carbon-11- and fluorine-18-radiolabeled pan-tropomyosin receptor kinase (Trk) inhibitors: exploration of the 4-aza-2-oxindole scaffold as Trk PET imaging agents. ACS Chem Neurosci 2015; 6:260-76. [PMID: 25350780 DOI: 10.1021/cn500193f] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Tropomyosin receptor kinases (TrkA/B/C) are critically involved in the development of the nervous system, in neurological disorders as well as in multiple neoplasms of both neural and non-neural origins. The development of Trk radiopharmaceuticals would offer unique opportunities toward a more complete understanding of this emerging therapeutic target. To that end, we first developed [(11)C]GW441756 ([(11)C]9), a high affinity photoisomerizable pan-Trk inhibitor, as a lead radiotracer for our positron emission tomography (PET) program. Efficient carbon-11 radiolabeling afforded [(11)C]9 in high radiochemical yields (isolated RCY, 25.9% ± 5.7%). In vitro autoradiographic studies in rat brain and TrkB-expressing human neuroblastoma cryosections confirmed that [(11)C]9 specifically binds to Trk receptors in vitro. MicroPET studies revealed that binding of [(11)C]9 in the rodent brain was mostly nonspecific despite initial high brain uptake (SUVmax = 2.0). Modeling studies of the 4-aza-2-oxindole scaffold led to the successful identification of a small series of high affinity fluorinated and methoxy derivatized pan-Trk inhibitors based on our lead compound 9. Out of this series, the fluorinated compound 10 was selected for initial evaluation and radiolabeled with fluorine-18 (isolated RCY, 2.5% ± 0.6%). Compound [(18)F]10 demonstrated excellent Trk selectivity in a panel of cancer relevant kinase targets and a promising in vitro profile in tumors and brain sections but high oxidative metabolic susceptibility leading to nonspecific brain distribution in vivo. The information gained in this study will guide further exploration of the 4-aza-2-oxindole scaffold as a lead for Trk PET ligand development.
Collapse
Affiliation(s)
- Vadim Bernard-Gauthier
- Experimental
Medicine, Department of Medicine, McGill University, 1110 Pine
Avenue West, Montreal, Quebec H3A 1A3, Canada
- Department
of Oncology, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 1Z2, Canada
| | - Arturo Aliaga
- Translational
Neuroimaging Laboratory, McGill Centre for Studies in Aging, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, Quebec H4H 1R3, Canada
| | - Antonio Aliaga
- McConnell
Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Mehdi Boudjemeline
- McConnell
Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Robert Hopewell
- McConnell
Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Alexey Kostikov
- McConnell
Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Pedro Rosa-Neto
- Translational
Neuroimaging Laboratory, McGill Centre for Studies in Aging, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, Quebec H4H 1R3, Canada
| | - Alexander Thiel
- Department
of Neurology and Neurosurgery, McGill University, Jewish General Hospital, 3755 Cote St. Catherine Rd., Montreal, Quebec H2T 1E2, Canada
| | - Ralf Schirrmacher
- Department
of Oncology, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 1Z2, Canada
- McConnell
Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| |
Collapse
|
46
|
Hirao K, Pontone GM, Smith GS. Molecular imaging of neuropsychiatric symptoms in Alzheimer's and Parkinson's disease. Neurosci Biobehav Rev 2015; 49:157-70. [PMID: 25446948 PMCID: PMC4806385 DOI: 10.1016/j.neubiorev.2014.11.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 10/27/2014] [Accepted: 11/12/2014] [Indexed: 01/14/2023]
Abstract
Neuropsychiatric symptoms (NPS) are very common in neurodegenerative diseases and are a major contributor to disability and caregiver burden. There is accumulating evidence that NPS may be a prodrome and/or a "risk factor" of neurodegenerative diseases. The medications used to treat these symptoms in younger patients are not very effective in patients with neurodegenerative disease and may have serious side effects. An understanding of the neurobiology of NPS is critical for the development of more effective intervention strategies. Targeting these symptoms may also have implications for prevention of cognitive or motor decline. Molecular brain imaging represents a bridge between basic and clinical observations and provides many opportunities for translation from animal models and human post-mortem studies to in vivo human studies. Molecular brain imaging studies in Alzheimer's disease (AD) and Parkinson's disease (PD) are reviewed with a primary focus on positron emission tomography studies of NPS. Future directions for the field of molecular imaging in AD and PD to understand the neurobiology of NPS will be discussed.
Collapse
Affiliation(s)
- Kentaro Hirao
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, USA; Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Gregory M Pontone
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, USA
| | - Gwenn S Smith
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, USA.
| |
Collapse
|
47
|
Snyder PJ, Lim YY, Schindler R, Ott BR, Salloway S, Daiello L, Getter C, Gordon CM, Maruff P. Microdosing of scopolamine as a "cognitive stress test": rationale and test of a very low dose in an at-risk cohort of older adults. Alzheimers Dement 2014; 10:262-7. [PMID: 24698030 DOI: 10.1016/j.jalz.2014.01.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Abnormal β-amyloid (Aβ) is associated with deleterious changes in central acetylcholinergic tone in the very early stages of Alzheimer's disease (AD), which may be unmasked by a cholinergic antagonist. We aimed to establish an optimal "microdose" of scopolamine for the development of a "cognitive stress test." METHODS Healthy older adults (n = 26, aged 55-75 years) with two risk factors for AD, but with low cortical Aβ burden, completed the Groton Maze Learning Test (GMLT) at baseline and then received scopolamine (0.20 mg subcutaneously). Participants were reassessed at 1, 3, 5, 7, and 8 hours postinjection. RESULTS There were significant differences, of a moderate magnitude, in performance between baseline and 3 hours postinjection for total errors, rule break errors, and the GMLT composite (d ≈ 0.50) that were all unrelated to body mass. CONCLUSIONS A very low dose of scopolamine leads to reliable cognitive impairment at 3 hours postdose (Tmax) and full cognitive recovery within 5 hours, supporting its use as a prognostic test paradigm to identify individuals with potential preclinical AD. This paradigm is being implemented in a larger cohort of healthy adults, with high or low Aβ, to identify pharmacodynamic differences between groups.
Collapse
Affiliation(s)
- Peter J Snyder
- Department of Neurology, Alpert Medical School, Brown University, Providence, RI, USA; Department of Neurology, Rhode Island Hospital, Providence, RI, USA.
| | - Yen Ying Lim
- Department of Neurology, Alpert Medical School, Brown University, Providence, RI, USA; Department of Neurology, Rhode Island Hospital, Providence, RI, USA
| | | | - Brian R Ott
- Department of Neurology, Alpert Medical School, Brown University, Providence, RI, USA; Department of Neurology, Rhode Island Hospital, Providence, RI, USA
| | - Stephen Salloway
- Department of Neurology, Alpert Medical School, Brown University, Providence, RI, USA; Department of Neurology, Butler Hospital, Providence, RI, USA
| | - Lori Daiello
- Department of Neurology, Alpert Medical School, Brown University, Providence, RI, USA; Department of Neurology, Rhode Island Hospital, Providence, RI, USA
| | - Christine Getter
- Lifespan Clinical Research Center, Rhode Island Hospital, Providence, RI, USA
| | - Catherine M Gordon
- Lifespan Clinical Research Center, Rhode Island Hospital, Providence, RI, USA; Department of Pediatrics, Alpert Medical School, Brown University, Providence, RI, USA
| | - Paul Maruff
- CogState, Ltd., Melbourne, Victoria, Australia; The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
48
|
Li X, Li M, Li Y, Quan Q, Wang J. Cellular and molecular mechanisms underlying the action of ginsenoside Rg1 against Alzheimer's disease. Neural Regen Res 2014; 7:2860-6. [PMID: 25317137 PMCID: PMC4190943 DOI: 10.3969/j.issn.1673-5374.2012.36.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 09/20/2012] [Indexed: 11/18/2022] Open
Abstract
Ginsenoside Rg1 inhibits oxidation, aging and cell apoptosis, and improves cognitive function. In this study, we pretreated rat brain tissue sections with ginsenoside Rg1, and established brain slice models of Alzheimer’s disease induced by okadaic acid. The results revealed that ginsenoside Rg1 pretreatment suppressed the increase in phosphorylated Tau protein expression induced by incubation with okadaic acid, and reduced brain-derived neurotrophic factor expression. These results suggest that ginsenoside Rg1 upregulates brain-derived neurotrophic factor expression and inhibits Tau protein phosphorylation in brain slices from a rat model of Alzheimer’s disease.
Collapse
Affiliation(s)
- Xi Li
- Department of Geriatrics, Second Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Ming Li
- Department of Geriatrics, Second Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Yuan Li
- Department of Encephalopathy, Xi'an Electric Power Central Hospital, Xi'an 710032, Shaanxi Province, China
| | - Qiankun Quan
- Research Center of Rehabilitation Science and Technology, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi Province, China
| | - Juan Wang
- Department of Laboratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| |
Collapse
|
49
|
Alldred MJ, Lee SH, Petkova E, Ginsberg SD. Expression profile analysis of vulnerable CA1 pyramidal neurons in young-Middle-Aged Ts65Dn mice. J Comp Neurol 2014; 523:61-74. [PMID: 25131634 DOI: 10.1002/cne.23663] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 08/07/2014] [Accepted: 08/07/2014] [Indexed: 12/19/2022]
Abstract
Down syndrome (DS) is the most prevalent cause of intellectual disability (ID). Individuals with DS show a variety of cognitive deficits, most notably in hippocampal learning and memory, and display pathological hallmarks of Alzheimer's disease (AD), with neurodegeneration of cholinergic basal forebrain (CBF) neurons. Elucidation of the molecular and cellular underpinnings of neuropathology has been assessed via gene expression analysis in a relevant animal model, termed the Ts65Dn mouse. The Ts65Dn mouse is a segmental trisomy model of DS that mimics DS/AD pathology, notably age-related cognitive dysfunction and degeneration of basal forebrain cholinergic neurons (BFCNs). To determine expression level changes, molecular fingerprinting of cornu ammonis 1 (CA1) pyramidal neurons was performed in adult (4-9 month-old) Ts65Dn mice, at the initiation of BFCN degeneration. To quantitate transcriptomic changes during this early time period, laser capture microdissection (LCM), terminal continuation (TC) RNA amplification, custom-designed microarray analysis, and subsequent validation of individual transcripts by qPCR and protein analysis via immunoblotting was performed. The results indicate significant alterations within CA1 pyramidal neurons of Ts65Dn mice compared with normal disomic (2N) littermates, notably in the downregulation of neurotrophins and their cognate neurotrophin receptors among other classes of transcripts relevant to neurodegeneration. The results of this single-population gene expression analysis at the time of septohippocampal deficits in a trisomic mouse model shed light on a vulnerable circuit that may cause the AD-like pathology invariably seen in DS that could help to identify mechanisms of degeneration, and provide novel gene targets for therapeutic interventions. J. Comp. Neurol. 523:61-74, 2015. © 2014 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Melissa J Alldred
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York, 10962; Department of Psychiatry, New York University Langone Medical Center, New York, New York, 10016
| | | | | | | |
Collapse
|
50
|
Alldred MJ, Lee SH, Petkova E, Ginsberg SD. Expression profile analysis of hippocampal CA1 pyramidal neurons in aged Ts65Dn mice, a model of Down syndrome (DS) and Alzheimer's disease (AD). Brain Struct Funct 2014; 220:2983-96. [PMID: 25031177 DOI: 10.1007/s00429-014-0839-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 07/02/2014] [Indexed: 11/29/2022]
Abstract
Down syndrome (DS) is caused by the triplication of human chromosome 21 (HSA21) and is the most common genetic cause of intellectual disability, with individuals having deficits in cognitive function including hippocampal learning and memory and neurodegeneration of cholinergic basal forebrain neurons, a pathological hallmark of Alzheimer's disease (AD). To date, the molecular underpinnings driving this pathology have not been elucidated. The Ts65Dn mouse is a segmental trisomy model of DS and like DS/AD pathology, displays age-related cognitive dysfunction and basal forebrain cholinergic neuron (BFCN) degeneration. To determine molecular and cellular changes important for elucidating mechanisms of neurodegeneration in DS/AD pathology, expression profiling studies were performed. Molecular fingerprinting of homogeneous populations of Cornu Ammonis 1 (CA1) pyramidal neurons was performed via laser capture microdissection followed by Terminal Continuation RNA amplification combined with custom-designed microarray analysis and subsequent validation of individual transcripts by qPCR and protein analysis via immunoblotting. Significant alterations were observed within CA1 pyramidal neurons of aged Ts65Dn mice compared to normal disomic (2N) littermates, notably in excitatory and inhibitory neurotransmission receptor families and neurotrophins, including brain-derived neurotrophic factor as well as several cognate neurotrophin receptors. Examining gene and protein expression levels after the onset of BFCN degeneration elucidated transcriptional and translational changes in neurons within a vulnerable circuit that may cause the AD-like pathology seen in DS as these individuals age, and provide rational targets for therapeutic interventions.
Collapse
Affiliation(s)
- Melissa J Alldred
- Center for Dementia Research, Nathan Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA
| | | | | | | |
Collapse
|