1
|
Trinh PNH, Baltos JA, Hellyer SD, May LT, Gregory KJ. Adenosine receptor signalling in Alzheimer’s disease. Purinergic Signal 2022; 18:359-381. [PMID: 35870032 PMCID: PMC9391555 DOI: 10.1007/s11302-022-09883-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/02/2022] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common dementia in the elderly and its increasing prevalence presents treatment challenges. Despite a better understanding of the disease, the current mainstay of treatment cannot modify pathogenesis or effectively address the associated cognitive and memory deficits. Emerging evidence suggests adenosine G protein-coupled receptors (GPCRs) are promising therapeutic targets for Alzheimer’s disease. The adenosine A1 and A2A receptors are expressed in the human brain and have a proposed involvement in the pathogenesis of dementia. Targeting these receptors preclinically can mitigate pathogenic β-amyloid and tau neurotoxicity whilst improving cognition and memory. In this review, we provide an accessible summary of the literature on Alzheimer’s disease and the therapeutic potential of A1 and A2A receptors. Although there are no available medicines targeting these receptors approved for treating dementia, we provide insights into some novel strategies, including allosterism and the targeting of oligomers, which may increase drug discovery success and enhance the therapeutic response.
Collapse
Affiliation(s)
- Phuc N. H. Trinh
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052 Australia
- Department of Pharmacology, Monash University, Parkville, VIC 3052 Australia
| | - Jo-Anne Baltos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052 Australia
- Department of Pharmacology, Monash University, Parkville, VIC 3052 Australia
| | - Shane D. Hellyer
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052 Australia
| | - Lauren T. May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052 Australia
- Department of Pharmacology, Monash University, Parkville, VIC 3052 Australia
| | - Karen J. Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052 Australia
- Department of Pharmacology, Monash University, Parkville, VIC 3052 Australia
- ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Parkville, 3052 Australia
| |
Collapse
|
2
|
Brain Glucose Transporters: Role in Pathogenesis and Potential Targets for the Treatment of Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22158142. [PMID: 34360906 PMCID: PMC8348194 DOI: 10.3390/ijms22158142] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/06/2021] [Accepted: 07/28/2021] [Indexed: 12/16/2022] Open
Abstract
The most common cause of dementia, especially in elderly people, is Alzheimer’s disease (AD), with aging as its main risk factor. AD is a multifactorial neurodegenerative disease. There are several factors increasing the risk of AD development. One of the main features of Alzheimer’s disease is impairment of brain energy. Hypometabolism caused by decreased glucose uptake is observed in specific areas of the AD-affected brain. Therefore, glucose hypometabolism and energy deficit are hallmarks of AD. There are several hypotheses that explain the role of glucose hypometabolism in AD, but data available on this subject are poor. Reduced transport of glucose into neurons may be related to decreased expression of glucose transporters in neurons and glia. On the other hand, glucose transporters may play a role as potential targets for the treatment of AD. Compounds such as antidiabetic drugs, agonists of SGLT1, insulin, siRNA and liposomes are suggested as therapeutics. Nevertheless, the suggested targets of therapy need further investigations.
Collapse
|
3
|
Lu J, Bao W, Li M, Li L, Zhang Z, Alberts I, Brendel M, Cumming P, Lu H, Xiao Z, Zuo C, Guan Y, Zhao Q, Rominger A. Associations of [ 18F]-APN-1607 Tau PET Binding in the Brain of Alzheimer's Disease Patients With Cognition and Glucose Metabolism. Front Neurosci 2020; 14:604. [PMID: 32694971 PMCID: PMC7338611 DOI: 10.3389/fnins.2020.00604] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/18/2020] [Indexed: 11/16/2022] Open
Abstract
Molecular imaging of tauopathies is complicated by the differing specificities and off-target binding properties of available radioligands for positron emission tomography (PET). [18F]-APN-1607 ([18F]-PM-PBB3) is a newly developed PET tracer with promising properties for tau imaging. We aimed to characterize the cerebral binding of [18F]-APN-1607 in Alzheimer's disease (AD) patients compared to normal control (NC) subjects. Therefore, we obtained static late frame PET recordings with [18F]-APN-1607 and [18F]-FDG in patients with a clinical diagnosis of AD group, along with an age-matched NC group ([18F]-APN-1607 only). Using statistical parametric mapping (SPM) and volume of interest (VOI) analyses of the reference region normalized standardized uptake value ratio maps, we then tested for group differences and relationships between both PET biomarkers, as well as their associations with clinical general cognition. In the AD group, [18F]-APN-1607 binding was elevated in widespread cortical regions (P < 0.001 for VOI analysis, familywise error-corrected P < 0.01 for SPM analysis). The regional uptake in AD patients correlated negatively with Mini-Mental State Examination score (frontal lobe: R = -0.632, P = 0.004; temporal lobe: R = -0.593, P = 0.008; parietal lobe: R = -0.552, P = 0.014; insula: R = -0.650, P = 0.003; cingulum: R = -0.665, P = 0.002) except occipital lobe (R = -0.417, P = 0.076). The hypometabolism to [18F]-FDG PET in AD patients also showed negative correlations with regional [18F]-APN-1607 binding in some signature areas of AD (temporal lobe: R = -0.530, P = 0.020; parietal lobe: R = -0.637, P = 0.003; occipital lobe: R = -0.567, P = 0.011). In conclusion, our results suggested that [18F]-APN-1607 PET sensitively detected tau deposition in AD and that individual tauopathy correlated with impaired cerebral glucose metabolism and cognitive function.
Collapse
Affiliation(s)
- Jiaying Lu
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Weiqi Bao
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ming Li
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ling Li
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhengwei Zhang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ian Alberts
- Department of Nuclear Medicine, University Hospital Bern, Bern, Switzerland
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Paul Cumming
- Department of Nuclear Medicine, University Hospital Bern, Bern, Switzerland
- Faculty of Health, School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD, Australia
| | - Huimeng Lu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhenxu Xiao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuantao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qianhua Zhao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Axel Rominger
- Department of Nuclear Medicine, University Hospital Bern, Bern, Switzerland
| |
Collapse
|
4
|
Rüb U, Stratmann K, Heinsen H, Seidel K, Bouzrou M, Korf HW. Alzheimer's Disease: Characterization of the Brain Sites of the Initial Tau Cytoskeletal Pathology Will Improve the Success of Novel Immunological Anti-Tau Treatment Approaches. J Alzheimers Dis 2017; 57:683-696. [PMID: 28269779 DOI: 10.3233/jad-161102] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) represents the most frequent neurodegenerative disease of the human brain worldwide. Currently practiced treatment strategies for AD only include some less effective symptomatic therapeutic interventions, which unable to counteract the disease course of AD. New therapeutic attempts aimed to prevent, reduce, or remove the extracellular depositions of the amyloid-β protein did not elicit beneficial effects on cognitive deficits or functional decline of AD. In view of the failure of these amyloid-β-based therapeutic trials and the close correlation between the brain pathology of the cytoskeletal tau protein and clinical AD symptoms, therapeutic attention has since shifted to the tau cytoskeletal protein as a novel drug target. The abnormal hyperphosphorylation and intraneuronal aggregation of this protein are early events in the evolution of the AD-related neurofibrillary pathology, and the brain spread of the AD-related tau aggregation pathology may possibly follow a corruptive protein templating and seeding-like mechanism according to the prion hypothesis. Accordingly, immunotherapeutic targeting of the tau aggregation pathology during the very early pre-tangle phase is currently considered to represent an effective and promising therapeutic approach for AD. Recent studies have shown that the initial immunoreactive tau aggregation pathology already prevails in several subcortical regions in the absence of any cytoskeletal changes in the cerebral cortex. Thus, it may be hypothesized that the subcortical brain regions represent the "port of entry" for the pathogenetic agent from which the disease ascends anterogradely as an "interconnectivity pathology".
Collapse
Affiliation(s)
- Udo Rüb
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe-University, Frankfurt/Main, Germany
| | - Katharina Stratmann
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe-University, Frankfurt/Main, Germany
| | - Helmut Heinsen
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany.,Department of Pathology, Ageing Brain Study Group, University of São Paulo Medical School, São Paulo, Brazil
| | - Kay Seidel
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe-University, Frankfurt/Main, Germany
| | - Mohamed Bouzrou
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe-University, Frankfurt/Main, Germany
| | - Horst-Werner Korf
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe-University, Frankfurt/Main, Germany
| |
Collapse
|
5
|
Abstract
Alzheimer's disease (AD) is a major form of senile dementia, characterized by progressive memory and neuronal loss combined with cognitive impairment. AD is the most common neurodegenerative disease worldwide, affecting one-fifth of those aged over 85 years. Recent therapeutic approaches have been strongly influenced by five neuropathological hallmarks of AD: acetylcholine deficiency, glutamate excitotoxicity, extracellular deposition of amyloid-β (Aβ plague), formation of intraneuronal neurofibrillary tangles (NTFs), and neuroinflammation. The lowered concentrations of acetylcholine (ACh) in AD result in a progressive and significant loss of cognitive and behavioral function. Current AD medications, memantine and acetylcholinesterase inhibitors (AChEIs) alleviate some of these symptoms by enhancing cholinergic signaling, but they are not curative. Since 2003, no new drugs have been approved for the treatment of AD. This article focuses on the current research in clinical trials targeting the neuropathological findings of AD including acetylcholine response, glutamate transmission, Aβ clearance, tau protein deposits, and neuroinflammation. These investigations include acetylcholinesterase inhibitors, agonists and antagonists of neurotransmitter receptors, β-secretase (BACE) or γ-secretase inhibitors, vaccines or antibodies targeting Aβ clearance or tau protein, as well as anti-inflammation compounds. Ongoing Phase III clinical trials via passive immunotherapy against Aβ peptides (crenezumab, gantenerumab, and aducanumab) seem to be promising. Using small molecules blocking 5-HT6 serotonin receptor (intepirdine), inhibiting BACE activity (E2609, AZD3293, and verubecestat), or reducing tau aggregation (TRx0237) are also currently in Phase III clinical trials. We here systemically review the findings from recent clinical trials to provide a comprehensive review of novel therapeutic compounds in the treatment and prevention of AD.
Collapse
Affiliation(s)
- Shih-Ya Hung
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung, 40402 Taiwan
- Division of Colorectal Surgery, China Medical University Hospital, Taichung, 40447 Taiwan
| | - Wen-Mei Fu
- Pharmacological Institute, College of Medicine, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei, 10051 Taiwan
| |
Collapse
|
6
|
Pawlowski M, Meuth SG, Duning T. Cerebrospinal Fluid Biomarkers in Alzheimer's Disease-From Brain Starch to Bench and Bedside. Diagnostics (Basel) 2017; 7:diagnostics7030042. [PMID: 28703785 PMCID: PMC5617942 DOI: 10.3390/diagnostics7030042] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 06/21/2017] [Accepted: 07/06/2017] [Indexed: 12/13/2022] Open
Abstract
Alzheimer’s disease is the most common cause of dementia. Over the last three decades, research has advanced dramatically and provided a detailed understanding of the molecular events underlying the pathogenesis of Alzheimer’s disease. In parallel, assays for the detection of biomarkers that reflect the typical Alzheimer’s disease-associated pathology have been developed and validated in myriads of clinical studies. Such biomarkers complement clinical diagnosis and improve diagnostic accuracy. The use of biomarkers will become even more important with the advent of disease-modifying therapies. Such therapies will likely be most beneficial when administered early in the disease course. Here, we summarise the development of the core Alzheimer’s disease cerebrospinal fluid biomarkers: amyloid-β and tau. We provide an overview of their role in cellular physiology and Alzheimer’s disease pathology, and embed their development as cerebrospinal fluid biomarkers into the historical context of Alzheimer’s disease research. Finally, we summarise recommendations for their use in clinical practice, and outline perspectives for novel cerebrospinal fluid candidate biomarkers.
Collapse
Affiliation(s)
- Matthias Pawlowski
- Department of Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, Münster 48149, Germany.
| | - Sven G Meuth
- Department of Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, Münster 48149, Germany.
| | - Thomas Duning
- Department of Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, Münster 48149, Germany.
| |
Collapse
|
7
|
Szablewski L. Glucose Transporters in Brain: In Health and in Alzheimer’s Disease. J Alzheimers Dis 2016; 55:1307-1320. [DOI: 10.3233/jad-160841] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
8
|
Seripa D, Solfrizzi V, Imbimbo BP, Daniele A, Santamato A, Lozupone M, Zuliani G, Greco A, Logroscino G, Panza F. Tau-directed approaches for the treatment of Alzheimer's disease: focus on leuco-methylthioninium. Expert Rev Neurother 2016; 16:259-77. [PMID: 26822031 DOI: 10.1586/14737175.2016.1140039] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Small molecular weight compounds able to inhibit formation of tau oligomers and fibrils have already been tested for Alzheimer's disease (AD) treatment. The most advanced tau aggregation inhibitor (TAI) is methylthioninium (MT), a drug existing in equilibrium between a reduced (leuco-methylthioninium) and oxidized form (MT(+)). MT chloride (also known as methylene blue) was investigated in a 24-week Phase II study in 321 mild-to-moderate AD patients at the doses of 69, 138, and 228 mg/day. This trial failed to show significant positive effects of MT in the overall patient population. The dose of 138 mg/day showed potential benefits on cognitive performance of moderately affected patients and cerebral blood flow in mildly affected patients. A follow-up compound (TRx0237) claimed to be more bioavailable and less toxic than MT, is now being developed. Phase III clinical trials on this novel TAI in AD and in the behavioral variant of frontotemporal dementia are underway.
Collapse
Affiliation(s)
- Davide Seripa
- a Geriatric Unit & Laboratory of Gerontology and Geriatrics, Department of Medical Sciences , IRCCS 'Casa Sollievo della Sofferenza' , San Giovanni Rotondo , Foggia , Italy
| | - Vincenzo Solfrizzi
- b Geriatric Medicine-Memory Unit and Rare Disease Centre , University of Bari Aldo Moro , Bari , Italy
| | - Bruno P Imbimbo
- c Research & Development Department , Chiesi Farmaceutici , Parma , Italy
| | - Antonio Daniele
- d Institute of Neurology , Catholic University of Sacred Heart , Rome , Italy
| | - Andrea Santamato
- e Physical Medicine and Rehabilitation Section, 'OORR' Hospital , University of Foggia , Foggia , Italy
| | - Madia Lozupone
- f Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs , University of Bari Aldo Moro , Bari , Italy
| | - Giovanni Zuliani
- g Department of Medical Science, Section of Internal and Cardiopulmonary Medicine , University of Ferrara
| | - Antonio Greco
- a Geriatric Unit & Laboratory of Gerontology and Geriatrics, Department of Medical Sciences , IRCCS 'Casa Sollievo della Sofferenza' , San Giovanni Rotondo , Foggia , Italy
| | - Giancarlo Logroscino
- f Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs , University of Bari Aldo Moro , Bari , Italy.,h Department of Clinical Research in Neurology , University of Bari Aldo Moro, 'Pia Fondazione Cardinale G. Panico' , Tricase , Lecce , Italy
| | - Francesco Panza
- a Geriatric Unit & Laboratory of Gerontology and Geriatrics, Department of Medical Sciences , IRCCS 'Casa Sollievo della Sofferenza' , San Giovanni Rotondo , Foggia , Italy.,f Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs , University of Bari Aldo Moro , Bari , Italy.,h Department of Clinical Research in Neurology , University of Bari Aldo Moro, 'Pia Fondazione Cardinale G. Panico' , Tricase , Lecce , Italy
| |
Collapse
|
9
|
Rüb U, Stratmann K, Heinsen H, Del Turco D, Ghebremedhin E, Seidel K, den Dunnen W, Korf HW. Hierarchical Distribution of the Tau Cytoskeletal Pathology in the Thalamus of Alzheimer's Disease Patients. J Alzheimers Dis 2016; 49:905-15. [PMID: 26519431 DOI: 10.3233/jad-150639] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In spite of considerable progress in neuropathological research on Alzheimer's disease (AD), knowledge regarding the exact pathoanatomical distribution of the tau cytoskeletal pathology in the thalamus of AD patients in the advanced Braak and Braak AD stages V or VI of the cortical cytoskeletal pathology is still fragmentary. Investigation of serial 100 μm-thick brain tissue sections through the thalamus of clinically diagnosed AD patients with Braak and Braak AD stage V or VI cytoskeletal pathologies immunostained with the anti-tau AT8 antibody, along with the affection of the extraterritorial reticular nucleus of the thalamus, reveals a consistent and severe tau immunoreactive cytoskeletal pathology in the limbic nuclei of the thalamus (e.g., paraventricular, anterodorsal and laterodorsal nuclei, limitans-suprageniculate complex). The thalamic nuclei integrated into the associative networks of the human brain (e.g., ventral anterior and mediodorsal nuclei) are only mildly affected, while its motor precerebellar (ventral lateral nucleus) and sensory nuclei (e.g., lateral and medial geniculate bodies, ventral posterior medial and lateral nuclei, parvocellular part of the ventral posterior medial nucleus) are more or less spared. The highly stereotypical and characteristic thalamic distribution pattern of the AD-related tau cytoskeletal pathology represents an anatomical mirror of the hierarchical topographic distribution of the cytoskeletal pathology in the interconnected regions of the cerebral cortex of AD patients. These pathoanatomical parallels support the pathophysiological concept of a transneuronal spread of the disease process of AD along anatomical pathways. The AD-related tau cytoskeletal pathology in the thalamus most likely contributes substantially to the neuropsychiatric disease symptoms (e.g., dementia), attention deficits, oculomotor dysfunctions, altered non-discriminative aspects of pain experience of AD patients, and the disruption of their waking and sleeping patterns.
Collapse
Affiliation(s)
- Udo Rüb
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe-University, Frankfurt/Main, Germany
| | - Katharina Stratmann
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe-University, Frankfurt/Main, Germany
| | - Helmut Heinsen
- Morphological Brain Research Unit, Psychiatric Clinic, Julius Maximilians University, Würzburg, Germany
| | - Domenico Del Turco
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University, Frankfurt/Main, Germany
| | - Estifanos Ghebremedhin
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University, Frankfurt/Main, Germany
| | - Kay Seidel
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe-University, Frankfurt/Main, Germany
| | - Wilfred den Dunnen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Horst-Werner Korf
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe-University, Frankfurt/Main, Germany
| |
Collapse
|
10
|
Tau-Centric Targets and Drugs in Clinical Development for the Treatment of Alzheimer's Disease. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3245935. [PMID: 27429978 PMCID: PMC4939203 DOI: 10.1155/2016/3245935] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 05/19/2016] [Indexed: 11/17/2022]
Abstract
The failure of several Phase II/III clinical trials in Alzheimer's disease (AD) with drugs targeting β-amyloid accumulation in the brain fuelled an increasing interest in alternative treatments against tau pathology, including approaches targeting tau phosphatases/kinases, active and passive immunization, and anti-tau aggregation. The most advanced tau aggregation inhibitor (TAI) is methylthioninium (MT), a drug existing in equilibrium between a reduced (leuco-methylthioninium) and oxidized form (MT+). MT chloride (methylene blue) was investigated in a 24-week Phase II clinical trial in 321 patients with mild to moderate AD that failed to show significant positive effects in mild AD patients, although long-term observations (50 weeks) and biomarker studies suggested possible benefit. The dose of 138 mg/day showed potential benefits on cognitive performance of moderately affected AD patients and cerebral blood flow in mildly affected patients. Further clinical evidence will come from the large ongoing Phase III trials for the treatment of AD and the behavioral variant of frontotemporal dementia on a new form of this TAI, more bioavailable and less toxic at higher doses, called TRx0237. More recently, inhibitors of tau acetylation are being actively pursued based on impressive results in animal studies obtained by salsalate, a clinically used derivative of salicylic acid.
Collapse
|
11
|
Stratmann K, Heinsen H, Korf HW, Del Turco D, Ghebremedhin E, Seidel K, Bouzrou M, Grinberg LT, Bohl J, Wharton SB, den Dunnen W, Rüb U. Precortical Phase of Alzheimer's Disease (AD)-Related Tau Cytoskeletal Pathology. Brain Pathol 2015; 26:371-86. [PMID: 26193084 DOI: 10.1111/bpa.12289] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/13/2015] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease (AD) represents the most frequent progressive neuropsychiatric disorder worldwide leading to dementia. We systematically investigated the presence and extent of the AD-related cytoskeletal pathology in serial thick tissue sections through all subcortical brain nuclei that send efferent projections to the transentorhinal and entorhinal regions in three individuals with Braak and Braak AD stage 0 cortical cytoskeletal pathology and fourteen individuals with Braak and Braak AD stage I cortical cytoskeletal pathology by means of immunostainings with the anti-tau antibody AT8. These investigations revealed consistent AT8 immunoreactive tau cytoskeletal pathology in a subset of these subcortical nuclei in the Braak and Braak AD stage 0 individuals and in all of these subcortical nuclei in the Braak and Braak AD stage I individuals. The widespread affection of the subcortical nuclei in Braak and Braak AD stage I shows that the extent of the early subcortical tau cytoskeletal pathology has been considerably underestimated previously. In addition, our novel findings support the concept that subcortical nuclei become already affected during an early 'pre-cortical' evolutional phase before the first AD-related cytoskeletal changes occur in the mediobasal temporal lobe (i.e. allocortical transentorhinal and entorhinal regions). The very early involved subcortical brain regions may represent the origin of the AD-related tau cytoskeletal pathology, from where the neuronal cytoskeletal pathology takes an ascending course toward the secondarily affected allocortex and spreads transneuronally along anatomical pathways in predictable sequences.
Collapse
Affiliation(s)
- Katharina Stratmann
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe University, Frankfurt/Main, Germany
| | - Helmut Heinsen
- Morphological Brain Research Unit, Psychiatric Clinic, Julius Maximilians University Würzburg, Würzburg, Germany
| | - Horst-Werner Korf
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe University, Frankfurt/Main, Germany
| | - Domenico Del Turco
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University, Frankfurt/Main, Germany
| | - Estifanos Ghebremedhin
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University, Frankfurt/Main, Germany
| | - Kay Seidel
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe University, Frankfurt/Main, Germany
| | - Mohamed Bouzrou
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe University, Frankfurt/Main, Germany
| | - Lea T Grinberg
- Department of Neurology, Memory and Aging Center, University of California at San Francisco, San Francisco, CA.,Department of Pathology, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Jürgen Bohl
- Neuropathology Division, University Clinic of Mainz, Mainz, Germany
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Wilfred den Dunnen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Udo Rüb
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe University, Frankfurt/Main, Germany
| |
Collapse
|
12
|
Camboni M, Wang CM, Miranda C, Yoon JH, Xu R, Zygmunt D, Kaspar BK, Martin PT. Active and passive immunization strategies based on the SDPM1 peptide demonstrate pre-clinical efficacy in the APPswePSEN1dE9 mouse model for Alzheimer's disease. Neurobiol Dis 2013; 62:31-43. [PMID: 24021662 DOI: 10.1016/j.nbd.2013.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 07/18/2013] [Accepted: 09/01/2013] [Indexed: 12/31/2022] Open
Abstract
Recent clinical and pre-clinical studies suggest that both active and passive immunization strategies targeting Aβ amyloid may have clinical benefit in Alzheimer's disease. Here, we demonstrate that vaccination of APPswePSEN1dE9 mice with SDPM1, an engineered non-native Aβ amyloid-specific binding peptide, lowers brain Aβ amyloid plaque burden and brain Aβ1-40 and Aβ1-42 peptide levels, improves cognitive learning and memory in Morris water maze tests and increases the expression of synaptic brain proteins. This was the case in young mice immunized prior to development of significant brain amyloid burden, and in older mice, where brain amyloid was already present. Active immunization was optimized using ALUM as an adjuvant to stimulate production of anti-SDPM1 and anti-Aβ amyloid antibodies. Intracerebral injection of P4D6, an SDPM1 peptide-mimotope antibody, also lowered brain amyloid plaque burden in APPswePSEN1dE9 mice. Additionally, P4D6 inhibited Aβ amyloid-mediated toxicity in cultured neuronal cells. The protein sequence of the variable domain within the P4D6 heavy chain was found to mimic a multimer of the SDPM1 peptide motif. These data demonstrate the efficacy of active and passive vaccine strategies to target Aβ amyloid oligomers using an engineered peptide-mimotope strategy.
Collapse
Affiliation(s)
- Marybeth Camboni
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA
| | - Chiou-Miin Wang
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA
| | - Carlos Miranda
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA
| | - Jung Hae Yoon
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA
| | - Rui Xu
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA
| | - Deborah Zygmunt
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA
| | - Brian K Kaspar
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA; Department of Pediatrics, The Ohio State University, USA; Department of Neuroscience, The Ohio State University, USA
| | - Paul T Martin
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA; Department of Pediatrics, The Ohio State University, USA; Department of Physiology and Cell Biology, The Ohio State University, USA.
| |
Collapse
|
13
|
Hyperdynamic microtubules, cognitive deficits, and pathology are improved in tau transgenic mice with low doses of the microtubule-stabilizing agent BMS-241027. J Neurosci 2012; 32:7137-45. [PMID: 22623658 DOI: 10.1523/jneurosci.0188-12.2012] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Tau is a microtubule (MT)-stabilizing protein that is altered in Alzheimer's disease (AD) and other tauopathies. It is hypothesized that the hyperphosphorylated, conformationally altered, and multimeric forms of tau lead to a disruption of MT stability; however, direct evidence is lacking in vivo. In this study, an in vivo stable isotope-mass spectrometric technique was used to measure the turnover, or dynamicity, of MTs in brains of living animals. We demonstrated an age-dependent increase in MT dynamics in two different tau transgenic mouse models, 3xTg and rTg4510. MT hyperdynamicity was dependent on tau expression, since a reduction of transgene expression with doxycycline reversed the MT changes. Treatment of rTg4510 mice with the epothilone, BMS-241027, also restored MT dynamics to baseline levels. In addition, MT stabilization with BMS-241027 had beneficial effects on Morris water maze deficits, tau pathology, and neurodegeneration. Interestingly, pathological and functional benefits of BMS-241027 were observed at doses that only partially reversed MT hyperdynamicity. Together, these data suggest that tau-mediated loss of MT stability may contribute to disease progression and that very low doses of BMS-241027 may be useful in the treatment of AD and other tauopathies.
Collapse
|
14
|
Panza F, Frisardi V, Solfrizzi V, Imbimbo BP, Logroscino G, Santamato A, Greco A, Seripa D, Pilotto A. Immunotherapy for Alzheimer's disease: from anti-β-amyloid to tau-based immunization strategies. Immunotherapy 2012; 4:213-38. [PMID: 22339463 DOI: 10.2217/imt.11.170] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The exact mechanisms leading to Alzheimer's disease (AD) are largely unknown, limiting the identification of effective disease-modifying therapies. The two principal neuropathological hallmarks of AD are extracellular β-amyloid (Aβ), peptide deposition (senile plaques) and intracellular neurofibrillary tangles containing hyperphosphorylated tau protein. During the last decade, most of the efforts of the pharmaceutical industry were directed against the production and accumulation of Aβ. The most innovative of the pharmacological approaches was the stimulation of Aβ clearance from the brain of AD patients via the administration of Aβ antigens (active vaccination) or anti-Aβ antibodies (passive vaccination). Several active and passive anti-Aβ vaccines are under clinical investigation. Unfortunately, the first active vaccine (AN1792, consisting of preaggregate Aβ and an immune adjuvant, QS-21) was abandoned because it caused meningoencephalitis in approximately 6% of treated patients. Anti-Aβ monoclonal antibodies (bapineuzumab and solanezumab) are now being developed. The clinical results of the initial studies with bapineuzumab were equivocal in terms of cognitive benefit. The occurrence of vasogenic edema after bapineuzumab, and more rarely brain microhemorrhages (especially in Apo E ε4 carriers), has raised concerns on the safety of these antibodies directed against the N-terminus of the Aβ peptide. Solanezumab, a humanized anti-Aβ monoclonal antibody directed against the midregion of the Aβ peptide, was shown to neutralize soluble Aβ species. Phase II studies showed a good safety profile of solanezumab, while studies on cerebrospinal and plasma biomarkers documented good signals of pharmacodynamic activity. Although some studies suggested that active immunization may be effective against tau in animal models of AD, very few studies regarding passive immunization against tau protein are currently available. The results of the large, ongoing Phase III trials with bapineuzumab and solanezumab will tell us if monoclonal anti-Aβ antibodies may slow down the rate of deterioration of AD. Based on the new diagnostic criteria of AD and on recent major failures of anti-Aβ drugs in mild-to-moderate AD patients, one could argue that clinical trials on potential disease-modifying drugs, including immunological approaches, should be performed in the early stages of AD.
Collapse
Affiliation(s)
- Francesco Panza
- Geriatric Unit & Gerontology-Geriatric Research Laboratory, IRCCS Casa Sollievo della Sofferenza, Foggia, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Costanza A, Weber K, Gandy S, Bouras C, Hof PR, Giannakopoulos P, Canuto A. Review: Contact sport-related chronic traumatic encephalopathy in the elderly: clinical expression and structural substrates. Neuropathol Appl Neurobiol 2012; 37:570-84. [PMID: 21696410 DOI: 10.1111/j.1365-2990.2011.01186.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Professional boxers and other contact sport athletes are exposed to repetitive brain trauma that may affect motor functions, cognitive performance, emotional regulation and social awareness. The term of chronic traumatic encephalopathy (CTE) was recently introduced to regroup a wide spectrum of symptoms such as cerebellar, pyramidal and extrapyramidal syndromes, impairments in orientation, memory, language, attention, information processing and frontal executive functions, as well as personality changes and behavioural and psychiatric symptoms. Magnetic resonance imaging usually reveals hippocampal and vermis atrophy, a cavum septum pellucidum, signs of diffuse axonal injury, pituitary gland atrophy, dilated perivascular spaces and periventricular white matter disease. Given the partial overlapping of the clinical expression, epidemiology and pathogenesis of CTE and Alzheimer's disease (AD), as well as the close association between traumatic brain injuries (TBIs) and neurofibrillary tangle formation, a mixed pathology promoted by pathogenetic cascades resulting in either CTE or AD has been postulated. Molecular studies suggested that TBIs increase the neurotoxicity of the TAR DNA-binding protein 43 (TDP-43) that is a key pathological marker of ubiquitin-positive forms of frontotemporal dementia (FTLD-TDP) associated or not with motor neurone disease/amyotrophic lateral sclerosis (ALS). Similar patterns of immunoreactivity for TDP-43 in CTE, FTLD-TDP and ALS as well as epidemiological correlations support the presence of common pathogenetic mechanisms. The present review provides a critical update of the evolution of the concept of CTE with reference to its neuropathological definition together with an in-depth discussion of the differential diagnosis between this entity, AD and frontotemporal dementia.
Collapse
Affiliation(s)
- A Costanza
- Department of Psychiatry, University Hospitals and Faculty of Medicine, University of Geneva School of Medicine, Geneva, Switzerland
| | | | | | | | | | | | | |
Collapse
|
16
|
Kumari U, Heese K. Cardiovascular dementia - a different perspective. Open Biochem J 2010; 4:29-52. [PMID: 20448820 PMCID: PMC2864432 DOI: 10.2174/1874091x01004010029] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2009] [Revised: 01/12/2010] [Accepted: 01/22/2010] [Indexed: 02/08/2023] Open
Abstract
The number of dementia patients has been growing in recent years and dementia represents a significant threat to aging people all over the world. Recent research has shown that the number of people affected by Alzheimer's disease (AD) and dementia is growing at an epidemic pace. The rapidly increasing financial and personal costs will affect the world's economies, health care systems, and many families. Researchers are now exploring a possible connection among AD, vascular dementia (VD), diabetes mellitus (type 2, T2DM) and cardiovascular diseases (CD). This correlation may be due to a strong association of cardiovascular risk factors with AD and VD, suggesting that these diseases share some biologic pathways. Since heart failure is associated with an increased risk of AD and VD, keeping the heart healthy may prove to keep the brain healthy as well. The risk for dementia is especially high when diabetes mellitus is comorbid with severe systolic hypertension or heart disease. In addition, the degree of coronary artery disease (CAD) is independently associated with cardinal neuropathological lesions of AD. Thus, the contribution of T2DM and CD to AD and VD implies that cardiovascular therapies may prove useful in preventing AD and dementia.
Collapse
Affiliation(s)
- Udhaya Kumari
- Division of Cell and Molecular Biology, School of Biological Sciences, College of Science, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | | |
Collapse
|
17
|
Azorsa DO, Robeson RH, Frost D, Meec hoovet B, Brautigam GR, Dickey C, Beaudry C, Basu GD, Holz DR, Hernandez JA, Bisanz KM, Gwinn L, Grover A, Rogers J, Reiman EM, Hutton M, Stephan DA, Mousses S, Dunckley T. High-content siRNA screening of the kinome identifies kinases involved in Alzheimer's disease-related tau hyperphosphorylation. BMC Genomics 2010; 11:25. [PMID: 20067632 PMCID: PMC2820455 DOI: 10.1186/1471-2164-11-25] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Accepted: 01/12/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neurofibrillary tangles (NFT), a cardinal neuropathological feature of Alzheimer's disease (AD) that is highly correlated with synaptic loss and dementia severity, appear to be partly attributable to increased phosphorylation of the microtubule stabilizing protein tau at certain AD-related residues. Identifying the kinases involved in the pathologic phosphorylation of tau may provide targets at which to aim new AD-modifying treatments. RESULTS We report results from a screen of 572 kinases in the human genome for effects on tau hyperphosphorylation using a loss of function, high-throughput RNAi approach. We confirm effects of three kinases from this screen, the eukaryotic translation initiation factor 2 alpha kinase 2 (EIF2AK2), the dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1A (DYRK1A), and the A-kinase anchor protein 13 (AKAP13) on tau phosphorylation at the 12E8 epitope (serine 262/serine 356). We provide evidence that EIF2AK2 effects may result from effects on tau protein expression, whereas DYRK1A and AKAP13 are likely more specifically involved in tau phosphorylation pathways. CONCLUSIONS These findings identify novel kinases that phosphorylate tau protein and provide a valuable reference data set describing the kinases involved in phosphorylating tau at an AD-relevant epitope.
Collapse
Affiliation(s)
- David O Azorsa
- Pharmaceutical Genomics Division, Translational Genomics Research Institute, Scottsdale, Arizona 85251, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Panza F, Solfrizzi V, Frisardi V, Imbimbo BP, Capurso C, D'Introno A, Colacicco AM, Seripa D, Vendemiale G, Capurso A, Pilotto A. Beyond the neurotransmitter-focused approach in treating Alzheimer's disease: drugs targeting beta-amyloid and tau protein. Aging Clin Exp Res 2009; 21:386-406. [PMID: 20154508 DOI: 10.1007/bf03327445] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Drugs currently used to treat Alzheimer's Disease (AD) have limited therapeutic value and do not affect the main neuropathological hallmarks of the disease, i.e., senile plaques and neurofibrillar tangles. Senile plaques are mainly formed of beta-amyloid (Abeta), a 42-aminoacid peptide. Neurofibrillar tangles are composed of paired helical filaments of hyperphosphorylated tau protein. New, potentially disease-modifying, therapeutic approaches are targeting Abeta and tau protein. Drugs directed against Abeta include active and passive immunization, that have been found to accelerate Abeta clearance from the brain. The most developmentally advanced monoclonal antibody directly targeting Abeta is bapineuzumab, now being studied in a large Phase III clinical trial. Compounds that interfere with proteases regulating Abeta formation from amyloid precursor protein (APP) are also actively pursued. The discovery of inhibitors of beta-secretase, the enzyme that regulates the first step of the amyloidogenic metabolism of APP, has been revealed to be particularly difficult due to inherent medicinal chemistry problems, and only one compound (CTS-21166) has reached clinical testing. Conversely, several compounds that inhibit gamma-secretase, the pivotal enzyme that generates Abeta, have been identified, the most advanced being LY-450139 (semagacestat), now in Phase III clinical development. Compounds that stimulate alpha-secretase, the enzyme responsible for the non-amyloidogenic metabolism of APP, are also being developed, and one of them, EHT-0202, has recently entered Phase II testing. Potent inhibitors of Abeta aggregation have also been identified, and one of such compounds, PBT-2, has provided encouraging neuropsychological results in a recently completed Phase II study. Therapeutic approaches directed against tau protein include inhibitors of glycogen synthase kinase- 3 (GSK-3), the enzyme responsible for tau phosphorylation and tau protein aggregation inhibitors. NP-12, a promising GSK-3 inhibitor, is being tested in a Phase II study, and methylthioninium chloride, a tau protein aggregation inhibitor, has given initial encouraging results in a 50-week study. With all these approaches on their way, the hope for disease-modifying therapy in this devastating disease may become a reality in the next 5 years.
Collapse
Affiliation(s)
- Francesco Panza
- Department of Geriatrics, Center for Aging Brain, Memory Unit, University of Bari, 70124, Bari, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Lace G, Savva GM, Forster G, de Silva R, Brayne C, Matthews FE, Barclay JJ, Dakin L, Ince PG, Wharton SB. Hippocampal tau pathology is related to neuroanatomical connections: an ageing population-based study. ACTA ACUST UNITED AC 2009; 132:1324-34. [PMID: 19321462 DOI: 10.1093/brain/awp059] [Citation(s) in RCA: 148] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Deposits of abnormally phosphorylated tau protein are found in numerous neurodegenerative disorders; the 'tauopathies', which include Alzheimer's and Pick's diseases, but tau pathology is also found in the ageing brain. Variation in tau pathology in brain ageing and its relationship to development of tauopathies and cognitive impairment remains unclear. We aimed to determine the extent and pattern of spread of tau pathology in the hippocampus, a susceptible region important in dementia and milder states of memory impairment, using hippocampal samples from the elderly population-based Medical Research Council Cognitive Function and Ageing Study neuropathology cohort. Tau deposition was assessed in hippocampal anatomical sub-regions using the AT8 antibody to phosphorylated tau and isoform-specific antibodies to 3 and 4-repeat tau (RD3 and RD4). Abeta pathology was also assessed. In this population sample, which includes the full ageing spectrum from individuals with no cognitive impairment to those with dementia satisfying clinico-pathology criteria for Alzheimer's disease, we have demonstrated a high prevalence at death of tau pathology. AT8, Abeta, RD3 and RD4 showed similar regional distribution and increased RD3 was noted in late-stage ghost tangles. Abeta was shown to be a poor explanatory variable for tau pathology. Tau deposition progressed in a hierarchical manner. Hippocampal input regions and projection zones (such as lateral entorhinal cortex, CA1/subiculum border and outer molecular layer of dentate) were initially affected, with anterograde progression though the hippocampal circuitry. Six hippocampal tau anatomical stages were defined, each linking projectionally to their adjacent stages, suggesting spread of tau malfunction through neuroanatomical pathways in hippocampal ageing. These stages were significantly associated with dementia, and may provide a clinically useful tool in the clinico-pathological assessment of dementia and mild cognitive impairment.
Collapse
Affiliation(s)
- G Lace
- Academic Unit of Pathology, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
The difference in gliosis induced by β-amyloid and Tau treatments in astrocyte cultures derived from senescence accelerated and normal mouse strains. Biogerontology 2009; 10:695-710. [DOI: 10.1007/s10522-009-9217-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Accepted: 02/03/2009] [Indexed: 12/27/2022]
|
21
|
Medina PMB, Worthen RJ, Forsberg LJ, Brenman JE. The actin-binding protein capulet genetically interacts with the microtubule motor kinesin to maintain neuronal dendrite homeostasis. PLoS One 2008; 3:e3054. [PMID: 18725959 PMCID: PMC2516187 DOI: 10.1371/journal.pone.0003054] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2008] [Accepted: 08/06/2008] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Neurons require precise cytoskeletal regulation within neurites, containing microtubule tracks for cargo transport in axons and dendrites or within synapses containing organized actin. Due to the unique architecture and specialized function of neurons, neurons are particularly susceptible to perturbation of the cytoskeleton. Numerous actin-binding proteins help maintain proper cytoskeletal regulation. METHODOLOGY/PRINCIPAL FINDINGS From a Drosophila forward genetic screen, we identified a mutation in capulet--encoding a conserved actin-binding protein--that causes abnormal aggregates of actin within dendrites. Through interaction studies, we demonstrate that simultaneous genetic inactivation of capulet and kinesin heavy chain, a microtubule motor protein, produces elongate cofilin-actin rods within dendrites but not axons. These rods resemble actin-rich structures induced in both mammalian neurodegenerative and Drosophila Alzheimer's models, but have not previously been identified by loss of function mutations in vivo. We further demonstrate that mitochondria, which are transported by Kinesin, have impaired distribution along dendrites in a capulet mutant. While Capulet and Cofilin may biochemically cooperate in certain circumstances, in neuronal dendrites they genetically antagonize each other. CONCLUSIONS/SIGNIFICANCE The present study is the first molecularly defined loss of function demonstration of actin-cofilin rods in vivo. This study suggests that simultaneous, seemingly minor perturbations in neuronal dendrites can synergize producing severe abnormalities affecting actin, microtubules and mitochondria/energy availability in dendrites. Additionally, as >90% of Alzheimer's and Parkinson's cases are sporadic this study suggests mechanisms by which multiple mutations together may contribute to neurodegeneration instead of reliance on single mutations to produce disease.
Collapse
Affiliation(s)
- Paul M. B. Medina
- Neuroscience Center, UNC Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Ryan J. Worthen
- Neuroscience Center, UNC Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Lawrence J. Forsberg
- Neuroscience Center, UNC Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Jay E. Brenman
- Neuroscience Center, UNC Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
- Department of Cell and Developmental Biology, UNC Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
22
|
Barten DM, Albright CF. Therapeutic strategies for Alzheimer's disease. Mol Neurobiol 2008; 37:171-86. [PMID: 18581273 DOI: 10.1007/s12035-008-8031-2] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2008] [Accepted: 05/27/2008] [Indexed: 12/22/2022]
Abstract
Therapeutic approaches for Alzheimer's disease (AD) are guided by four disease characteristics: amyloid plaques, neurofibrillar tangles (NFT), neurodegeneration, and dementia. Amyloid plaques are composed largely of 4 kDa beta-amyloid (Abeta) peptides, with the more amyloidogenic, 42 amino acid form (Abeta42) as the primary species. Because multiple, rare mutations that cause early-onset, familial AD lead to increased production or aggregation of Abeta42, amyloid therapeutics aim to reduce the amount of toxic Abeta42 aggregates. Amyloid-based therapies include gamma-secretase inhibitors and modulators, BACE inhibitors, aggregation blockers, catabolism inducers, and anti-Abeta biologics. Tangles are composed of paired helical filaments of hyperphosphorylated tau protein. Tau-based therapeutics include kinase inhibitors, microtubule stabilizers, and catabolism inducers. Therapeutic strategies for neurodegeneration target multiple mechanisms, including excitotoxicity, mitochondrial dysfunction, oxidative damage, and inflammation or stimulation of neuronal viability. Although not disease modifying, cognition enhancers are important to treat the symptom of dementia. Strategies for cognition enhancement include cholinesterase inhibitors, and other approaches to enhance the signaling of cholinergic and glutamatergic neurons. In summary, plaques, tangles, neurodegeneration and dementia guide the development of multiple therapeutic approaches for AD and are the subject of this review.
Collapse
Affiliation(s)
- Donna M Barten
- Bristol Myers Squibb, Neuroscience Drug Discovery, 5 Research Parkway, Wallingford, CT 06492, USA.
| | | |
Collapse
|
23
|
Vershinin M, Xu J, Razafsky DS, King SJ, Gross SP. Tuning microtubule-based transport through filamentous MAPs: the problem of dynein. Traffic 2008; 9:882-92. [PMID: 18373727 DOI: 10.1111/j.1600-0854.2008.00741.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We recently proposed that regulating the single-to-multiple motor transition was a likely strategy for regulating kinesin-based transport in vivo. In this study, we use an in vitro bead assay coupled with an optical trap to investigate how this proposed regulatory mechanism affects dynein-based transport. We show that tau's regulation of kinesin function can proceed without interfering with dynein-based transport. Surprisingly, at extremely high tau levels--where kinesin cannot bind microtubules (MTs)--dynein can still contact MTs. The difference between tau's effects on kinesin- and dynein-based motility suggests that tau can be used to tune relative amounts of plus-end and minus-end-directed transport. As in the case of kinesin, we find that the 3RS isoform of tau is a more potent inhibitor of dynein binding to MTs. We show that this isoform-specific effect is not because of steric interference of tau's projection domains but rather because of tau's interactions with the motor at the MT surface. Nonetheless, we do observe a modest steric interference effect of tau away from the MT and discuss the potential implications of this for molecular motor structure.
Collapse
Affiliation(s)
- Michael Vershinin
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA
| | | | | | | | | |
Collapse
|
24
|
Farooqui T. Octopamine-mediated neuronal plasticity in honeybees: implications for olfactory dysfunction in humans. Neuroscientist 2007; 13:304-22. [PMID: 17644763 DOI: 10.1177/10738584070130040501] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Biogenic amines, such as norepinephrine (in vertebrates) and octopamine (in invertebrates), have structural and functional similarities. These amines play crucial roles in animal behavior by modifying the synaptic output of relevant neurons. Increased levels of norepinephrine in the olfactory bulb preferentially increase mitral cell excitatory responses to olfactory nerve inputs, suggesting its critical role in modulating olfactory function including memory formation and/or recall of specific olfactory memories. Increased levels of octopamine in the antennal lobe play an important role in a reinforcement pathway involved in olfactory learning and memory in honeybees. Similar to adrenergic receptors in the human brain, activation of octopaminergic receptors in the honeybee brain induces specific second messenger pathways that change protein phosphorylation and/or gene expression, altering the activity and/or abundance of proteins responsible for neuronal signaling leading to changes in olfactory behavior. The author's studies in honeybees Apis mellifera indicate that oxidative stress plays a major role in olfactory dysfunction. A similar mechanism has been proposed for olfactory abnormalities in patients of Alzheimer disease and Parkinson disease. Due to similarities in cellular and molecular processes, which govern neuronal plasticity in humans and honeybees, the author proposes that the honeybee can be used as a potential and relatively simple model system for understanding human olfactory dysfunction during aging and in neurodegenerative diseases.
Collapse
Affiliation(s)
- Tahira Farooqui
- Department of Entomology, The Ohio State University, Columbus, Ohio 43210, USA.
| |
Collapse
|
25
|
Areza-Fegyveres R, Rosemberg S, Castro RMR, Porto CS, Bahia VS, Caramelli P, Nitrini R. Dementia Pugilistica with clinical features of Alzheimer's disease. ARQUIVOS DE NEURO-PSIQUIATRIA 2007; 65:830-3. [DOI: 10.1590/s0004-282x2007000500019] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2007] [Accepted: 06/15/2007] [Indexed: 11/21/2022]
Abstract
A 61-year-old ex-boxer presented with a three-year history of progressive memory decline. During a seven-year follow-up period, there was a continuous cognitive decline, very similar to that usually observed in Alzheimer's disease. Parkinsonian, pyramidal or cerebellar signs were conspicuously absent. Neuropathological examination revealed the typical features of dementia pugilistica: cavum septi pellucidi with multiple fenestrations, numerous neurofibrillary tangles in the cerebral isocortex and hippocampus (and rare senile plaques). Immunohistochemistry disclosed a high number of tau protein deposits and scarce beta-amyloid staining. This case shows that dementia pugilistica may present with clinical features practically undistinguishable from Alzheimer's disease.
Collapse
|