1
|
Young RE, Zuccaro MV, LeDuc CA, Germain ND, Kim TH, Sarmiere P, Chung WK. Pathogenic PPP2R5D variants disrupt neuronal development and neurite outgrowth in patient-derived neurons that are reversed by allele-specific knockdown. HGG ADVANCES 2025; 6:100450. [PMID: 40340253 DOI: 10.1016/j.xhgg.2025.100450] [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: 06/06/2024] [Revised: 05/05/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025] Open
Abstract
A significant barrier to the treatment of neurodevelopmental disorders (NDDs) is a limited understanding of disease mechanisms. Heterozygous missense variants in PPP2R5D cause Houge-Janssens syndrome 1, a rare NDD characterized by macrocephaly, developmental delay, intellectual disability, seizures, autism spectrum disorder, and early-onset Parkinson disease. This study investigated the impact of pathogenic PPP2R5D variants on neuronal development and evaluated allele-specific knockdown as a potential therapeutic strategy. Induced pluripotent stem cells derived from individuals carrying the E198K and E420K variants, along with CRISPR-corrected isogenic controls, were differentiated into neural progenitors and cortical glutamatergic neurons. Patient-derived neural progenitors were hyper-proliferative, and glutamatergic neurons differentiated from these cells exhibited increased neurite outgrowth. Notably, neuronal overgrowth phenotypes were not observed in neurons lacking PPP2R5D, suggesting the disorder does not result from loss of function. RNA sequencing (RNA-seq) of glutamatergic neurons derived from patient lines compared to their isogenic controls revealed disruptions in pathways critical for neuronal development, synaptic signaling, and axon guidance. To target pathogenic transcripts, antisense oligonucleotides (ASOs) were designed to selectively knock down the E198K allele, the most common disease-causing missense variant. The most effective ASOs reversed neurite outgrowth defects in patient-derived neurons. These findings uncover molecular mechanisms underlying PPP2R5D-related NDDs and support allele-specific knockdown as a potential therapeutic approach.
Collapse
Affiliation(s)
- Randee E Young
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Pediatrics, Columbia University Medical Center, New York, NY 10032, USA
| | - Michael V Zuccaro
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Pediatrics, Columbia University Medical Center, New York, NY 10032, USA
| | - Charles A LeDuc
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Pediatrics, Columbia University Medical Center, New York, NY 10032, USA
| | | | - Tae Hyun Kim
- Ovid Therapeutics, Inc., New York, NY 10001, USA
| | | | - Wendy K Chung
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Pediatrics, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
2
|
Kovacs GG, Katsumata Y, Wu X, Aung KZ, Fardo DW, Forrest SL, Nelson PT. Amyloid-β predominant Alzheimer's disease neuropathologic change. Brain 2025; 148:401-407. [PMID: 39417691 PMCID: PMC11788189 DOI: 10.1093/brain/awae325] [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: 07/24/2024] [Revised: 08/22/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
Different subsets of Alzheimer's disease neuropathologic change (ADNC), including the intriguing set of individuals with severe/widespread amyloid-β (Aβ) plaques but no/mild tau tangles [Aβ-predominant (AP)-ADNC], may have distinct genetic and clinical features. Analysing National Alzheimer's Coordinating Center data, we stratified 1187 participants into AP-ADNC (n = 95), low Braak primary age-related tauopathy (PART; n = 185), typical-ADNC (n = 832) and high-Braak PART (n = 75). AP-ADNC differed in some clinical features and genetic polymorphisms in the APOE, SNX1, WNT3/MAPT and IGH genes. We conclude that AP-ADNC differs from classical ADNC with implications for in vivo studies.
Collapse
Affiliation(s)
- Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5T 0S8, Canada
- Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Yuriko Katsumata
- Department of Biostatistics, University of Kentucky, Lexington, KY 40536-0679, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Xian Wu
- Department of Biostatistics, University of Kentucky, Lexington, KY 40536-0679, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Khine Zin Aung
- Department of Biostatistics, University of Kentucky, Lexington, KY 40536-0679, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - David W Fardo
- Department of Biostatistics, University of Kentucky, Lexington, KY 40536-0679, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Shelley L Forrest
- Tanz Centre for Research in Neurodegenerative Disease and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5T 0S8, Canada
- Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
- Department of Pathology, Division of Neuropathology, University of Kentucky, Lexington, KY 40536-0679, USA
| |
Collapse
|
3
|
Tang R, Elman JA, Reynolds CA, Puckett OK, Panizzon MS, Lyons MJ, Hagler DJ, Fennema-Notestine C, Eyler LT, Dorros SM, Dale AM, Kremen WS, Franz CE. Cortical Surface Area Profile Mediates Effects of Childhood Disadvantage on Later-Life General Cognitive Ability. J Gerontol B Psychol Sci Soc Sci 2024; 79:gbae170. [PMID: 39383177 PMCID: PMC11561397 DOI: 10.1093/geronb/gbae170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Indexed: 10/11/2024] Open
Abstract
OBJECTIVES Childhood disadvantage is associated with lower general cognitive ability (GCA) and brain structural differences in midlife and older adulthood. However, the neuroanatomical mechanisms underlying childhood disadvantage effects on later-life GCA remain poorly understood. Although total surface area (SA) has been linked to lifespan GCA differences, total SA does not capture the nonuniform nature of childhood disadvantage effects on neuroanatomy, which varies across unimodal and transmodal cortices. Here, we examined whether cortical SA profile-the extent to which the spatial patterning of SA deviates from the normative unimodal-transmodal cortical organization-is a mediator of childhood disadvantage effects on later-life GCA. METHODS In 477 community-dwelling men aged 56-72 years old, childhood disadvantage index was derived from four indicators of disadvantages and GCA was assessed using a standardized test. Cortical SA was obtained from structural magnetic resonance imaging. For cortical SA profile, we calculated the spatial similarity between maps of individual cortical SA and MRI-derived principal gradient (i.e., unimodal-transmodal organization). Mediation analyses were conducted to examine the indirect effects of childhood disadvantage index through cortical SA profile on GCA. RESULTS Around 1.31% of childhood disadvantage index effects on later-life GCA were mediated by cortical SA profile, whereas total SA did not. Higher childhood disadvantage index was associated with more deviation of the cortical SA spatial patterning from the principal gradient, which in turn related to lower later-life GCA. DISCUSSION Childhood disadvantage may contribute to later-life GCA differences partly by influencing the spatial patterning of cortical SA in a way that deviates from the normative cortical organizational principle.
Collapse
Affiliation(s)
- Rongxiang Tang
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas, USA
| | - Jeremy A Elman
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California, USA
| | - Chandra A Reynolds
- Department of Psychology and Neuroscience, Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, Colorado, USA
| | - Olivia K Puckett
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California, USA
| | - Matthew S Panizzon
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California, USA
| | - Michael J Lyons
- Department of Psychological and Brain Sciences, Boston University, Boston, Massachusetts, USA
| | - Donald J Hagler
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Christine Fennema-Notestine
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Lisa T Eyler
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
- Desert Pacific Mental Illness Research Education and Clinical Center, VA San Diego Healthcare System, San Diego, California, USA
| | - Stephen M Dorros
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Anders M Dale
- Department of Radiology, University of California San Diego, La Jolla, California, USA
- Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - William S Kremen
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California, USA
| | - Carol E Franz
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
4
|
Aleksandrova EP, Ivlev AP, Kulikov AA, Naumova AA, Glazova MV, Chernigovskaya EV. Aging of Krushinsky-Molodkina audiogenic rats is accompanied with pronounced neurodegeneration and dysfunction of the glutamatergic system in the hippocampus. Brain Res 2024; 1846:149294. [PMID: 39461667 DOI: 10.1016/j.brainres.2024.149294] [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: 07/15/2024] [Revised: 09/30/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
Advancing age strongly correlates with an increased risk of epilepsy development. On the other hand, epilepsy may exacerbate the negative effects of aging making it pathological. In turn, the possible link between aging and epileptogenesis is dysregulation of glutamatergic transmission. In the present study, we analyzed the functional state of the glutamatergic system in the hippocampus of aging (18-month-old) Krushinsky-Molodkina (KM) audiogenic rats to disclose alterations associated with aging on the background of inherited predisposition to audiogenic seizures (AGS). Naïve KM rats with no AGS experience were recruited in the experiments. Wistar rats of the corresponding age were used as a control. First of all, aging KM rats demonstrated a significant decrease in cell population and synaptopodin expression in the hippocampus indicating enhanced loss of cells and synapses. Meanwhile, elevated phosphorylation of ERK1/2 and CREB and increased glutamate in the neuronal perikarya were revealed indicating increased activity of the rest hippocampal cells and increased glutamate production. However, glutamate in the fibers and synapses was mainly unchanged, and the proteins regulating glutamate exocytosis showed variable changes which could compensate each other and maintain glutamate release at the unchanged level. In addition, we revealed downregulation of NMDA-receptor subunit GluN2B and upregulation of AMPA-receptor GluA2 subunit, which could also prevent overexcitation and support cell survival in the hippocampus of aging KM rats. Nevertheless, abnormally high glutamate production, observed in aging KM rats, may provide the basis for hyperexcitability of the hippocampus and increased seizure susceptibility in old age.
Collapse
Affiliation(s)
- Ekaterina P Aleksandrova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, the Russian Academy of Sciences, St. Petersburg, Russian Federation.
| | - Andrey P Ivlev
- Sechenov Institute of Evolutionary Physiology and Biochemistry, the Russian Academy of Sciences, St. Petersburg, Russian Federation.
| | - Alexey A Kulikov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, the Russian Academy of Sciences, St. Petersburg, Russian Federation.
| | - Alexandra A Naumova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, the Russian Academy of Sciences, St. Petersburg, Russian Federation.
| | - Margarita V Glazova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, the Russian Academy of Sciences, St. Petersburg, Russian Federation.
| | - Elena V Chernigovskaya
- Sechenov Institute of Evolutionary Physiology and Biochemistry, the Russian Academy of Sciences, St. Petersburg, Russian Federation.
| |
Collapse
|
5
|
Brown RE. Measuring the replicability of our own research. J Neurosci Methods 2024; 406:110111. [PMID: 38521128 DOI: 10.1016/j.jneumeth.2024.110111] [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: 01/21/2024] [Revised: 03/08/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024]
Abstract
In the study of transgenic mouse models of neurodevelopmental and neurodegenerative disorders, we use batteries of tests to measure deficits in behaviour and from the results of these tests, we make inferences about the mental states of the mice that we interpret as deficits in "learning", "memory", "anxiety", "depression", etc. This paper discusses the problems of determining whether a particular transgenic mouse is a valid mouse model of disease X, the problem of background strains, and the question of whether our behavioural tests are measuring what we say they are. The problem of the reliability of results is then discussed: are they replicable between labs and can we replicate our results in our own lab? This involves the study of intra- and inter- experimenter reliability. The variables that influence replicability and the importance of conducting a complete behavioural phenotype: sensory, motor, cognitive and social emotional behaviour are discussed. Then the thorny question of failure to replicate is examined: Is it a curse or a blessing? Finally, the role of failure in research and what it tells us about our research paradigms is examined.
Collapse
Affiliation(s)
- Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
6
|
Walhovd KB, Krogsrud SK, Amlien IK, Sørensen Ø, Wang Y, Bråthen ACS, Overbye K, Kransberg J, Mowinckel AM, Magnussen F, Herud M, Håberg AK, Fjell AM, Vidal-Pineiro D. Fetal influence on the human brain through the lifespan. eLife 2024; 12:RP86812. [PMID: 38602745 PMCID: PMC11008813 DOI: 10.7554/elife.86812] [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] [Indexed: 04/12/2024] Open
Abstract
Human fetal development has been associated with brain health at later stages. It is unknown whether growth in utero, as indexed by birth weight (BW), relates consistently to lifespan brain characteristics and changes, and to what extent these influences are of a genetic or environmental nature. Here we show remarkably stable and lifelong positive associations between BW and cortical surface area and volume across and within developmental, aging and lifespan longitudinal samples (N = 5794, 4-82 y of age, w/386 monozygotic twins, followed for up to 8.3 y w/12,088 brain MRIs). In contrast, no consistent effect of BW on brain changes was observed. Partly environmental effects were indicated by analysis of twin BW discordance. In conclusion, the influence of prenatal growth on cortical topography is stable and reliable through the lifespan. This early-life factor appears to influence the brain by association of brain reserve, rather than brain maintenance. Thus, fetal influences appear omnipresent in the spacetime of the human brain throughout the human lifespan. Optimizing fetal growth may increase brain reserve for life, also in aging.
Collapse
Affiliation(s)
- Kristine B Walhovd
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
- Computational Radiology and Artificial Intelligence, Department of Radiology and Nuclear Medicine, Oslo University HospitalOsloNorway
| | - Stine K Krogsrud
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
| | - Inge K Amlien
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
| | - Øystein Sørensen
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
| | - Yunpeng Wang
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
| | | | - Knut Overbye
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
| | - Jonas Kransberg
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
| | | | - Fredrik Magnussen
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
| | - Martine Herud
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
| | - Asta K Håberg
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and TechnologyOsloNorway
| | - Anders Martin Fjell
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
- Computational Radiology and Artificial Intelligence, Department of Radiology and Nuclear Medicine, Oslo University HospitalOsloNorway
| | - Didac Vidal-Pineiro
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
| |
Collapse
|
7
|
Rudnitskaya E, Kozlova T, Burnyasheva A, Peunov D, Tyumentsev M, Stefanova N, Kolosova N. Postnatal Maturation of the Blood-Brain Barrier in Senescence-Accelerated OXYS Rats, Which Are Prone to an Alzheimer's Disease-like Pathology. Int J Mol Sci 2023; 24:15649. [PMID: 37958635 PMCID: PMC10648128 DOI: 10.3390/ijms242115649] [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: 09/21/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Alzheimer's disease (AD) is an old-age neurodegenerative disorder; however, AD predisposition may arise early in life. Vascular dysfunction makes a big contribution to AD development. Nonetheless, the possible role of early-life vascular dysfunction in AD development is still poorly investigated. Here, using OXYS rats as a suitable model of the most common (sporadic) type of AD, we investigated maturation of the blood-brain barrier (BBB) in the hippocampus and frontal cortex in the first 3 weeks of life. Using RNA-Seq data, we found an altered expression of BBB-associated genes in the middle of the first and second weeks of life in OXYS rats compared to control rats (Wistar strain). Moreover, by immunohistochemistry and electronic microscopy, we revealed a delay of vascularization and of subsequent pericyte coating of blood vessels in OXYS rats. These specific features were accompanied by an accelerated decrease in BBB permeability estimated using Evans blue dye. Notably, almost all of the observed differences from Wistar rats disappeared on postnatal day 20. Nonetheless, the observed features, which are characteristic of the postnatal period, may have long-term consequences and contribute to neurovascular dysfunction observed in OXYS rats late in life, thereby promoting early development of AD signs.
Collapse
Affiliation(s)
- Ekaterina Rudnitskaya
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (ICG SB RAS), 10 Lavrentyeva Ave., 630090 Novosibirsk, Russia; (T.K.); (A.B.); (D.P.); (M.T.); (N.S.); (N.K.)
| | | | | | | | | | | | | |
Collapse
|
8
|
Vasconcelos RO, Gordillo-Martinez F, Ramos A, Lau IH. Effects of Noise Exposure and Ageing on Anxiety and Social Behaviour in Zebrafish. BIOLOGY 2023; 12:1165. [PMID: 37759565 PMCID: PMC10525370 DOI: 10.3390/biology12091165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/11/2023] [Accepted: 08/11/2023] [Indexed: 09/29/2023]
Abstract
Noise pollution is creating a wide range of health problems related to physiological stress and anxiety that impact the social life of vertebrates, including humans. Ageing is known to be associated with changes in susceptibility to acoustic stimuli; however, the interaction between noise effects and senescence is not well understood. We tested the effects of 24 h continuous white noise (150 dB re 1 Pa) on both young adults and old zebrafish in terms of anxiety (novel tank diving test), social interactions (with mirror/conspecific attraction), and shoaling behaviour. Both noise and ageing induced higher anxiety responses in a novel environment. Since the old zebrafish showed longer bottom dwelling, acoustic treatment induced the opposite pattern with an initial increase in vertical exploration in the aged individuals. Both noise- and age-related anxiety responses were lowered when individuals were tested within a group. Regarding social interactions, both noise and ageing seemed to cause an increase in their proximity to a mirror. Although the results were not statistically significant, noise exposure seemed to further enhance conspecific attraction. Moreover, the interindividual distance within a shoal decreased with noise treatment in the aged individuals. This study is a first attempt to investigate the effects of both noise and ageing on zebrafish behaviour, suggesting the age-dependent physiological coping mechanisms associated with environmental stress.
Collapse
Affiliation(s)
- Raquel O. Vasconcelos
- Institute of Science and Environment, University of Saint Joseph, Macao, China
- MARE–Marine and Environmental Sciences Centre/ARNET—Aquatic Research Network, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal
- EPCV–Department of Life Sciences, Lusófona University, 1749-024 Lisbon, Portugal
| | | | - Andreia Ramos
- Institute of Science and Environment, University of Saint Joseph, Macao, China
| | - Ieng Hou Lau
- Institute of Science and Environment, University of Saint Joseph, Macao, China
| |
Collapse
|
9
|
Stefanova NA, Kolosova NG. The Rat Brain Transcriptome: From Infancy to Aging and Sporadic Alzheimer's Disease-like Pathology. Int J Mol Sci 2023; 24:ijms24021462. [PMID: 36674977 PMCID: PMC9865438 DOI: 10.3390/ijms24021462] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
It has been suggested that functional traits of the adult brain-all of which are established early in life-may affect the brain's susceptibility to Alzheimer's disease (AD). Results of our previous studies on senescence-accelerated OXYS rats, a model of sporadic AD, support this hypothesis. Here, to elucidate the molecular genetic nature of the aberrations revealed during brain maturation, we analyzed transcriptomes (RNA-seq data) of the prefrontal cortex (PFC) and hippocampus of OXYS rats and Wistar (control) rats in the period of brain maturation critical for OXYS rats (ages P3 and P10; P: postnatal day). We found more than 1000 differentially expressed genes in both brain structures; functional analysis indicated reduced efficiency of the formation of neuronal contacts, presumably explained mainly by deficits of mitochondrial functions. Next, we compared differentially expressed genes in the rat PFC and hippocampus from infancy to the progressive stage of AD-like pathology (five ages in total). Three genes (Thoc3, Exosc8, and Smpd4) showed overexpression in both brain regions of OXYS rats throughout the lifespan. Thus, reduced efficiency of the formation of neural networks in the brain of OXYS rats in infancy likely contributes to the development of their AD-like pathology.
Collapse
|
10
|
Grünblatt E, Homolak J, Babic Perhoc A, Davor V, Knezovic A, Osmanovic Barilar J, Riederer P, Walitza S, Tackenberg C, Salkovic-Petrisic M. From attention-deficit hyperactivity disorder to sporadic Alzheimer's disease-Wnt/mTOR pathways hypothesis. Front Neurosci 2023; 17:1104985. [PMID: 36875654 PMCID: PMC9978448 DOI: 10.3389/fnins.2023.1104985] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/31/2023] [Indexed: 02/18/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder with the majority of patients classified as sporadic AD (sAD), in which etiopathogenesis remains unresolved. Though sAD is argued to be a polygenic disorder, apolipoprotein E (APOE) ε4, was found three decades ago to pose the strongest genetic risk for sAD. Currently, the only clinically approved disease-modifying drugs for AD are aducanumab (Aduhelm) and lecanemab (Leqembi). All other AD treatment options are purely symptomatic with modest benefits. Similarly, attention-deficit hyperactivity disorder (ADHD), is one of the most common neurodevelopmental mental disorders in children and adolescents, acknowledged to persist in adulthood in over 60% of the patients. Moreover, for ADHD whose etiopathogenesis is not completely understood, a large proportion of patients respond well to treatment (first-line psychostimulants, e.g., methylphenidate/MPH), however, no disease-modifying therapy exists. Interestingly, cognitive impairments, executive, and memory deficits seem to be common in ADHD, but also in early stages of mild cognitive impairment (MCI), and dementia, including sAD. Therefore, one of many hypotheses is that ADHD and sAD might have similar origins or that they intercalate with one another, as shown recently that ADHD may be considered a risk factor for sAD. Intriguingly, several overlaps have been shown between the two disorders, e.g., inflammatory activation, oxidative stress, glucose and insulin pathways, wingless-INT/mammalian target of rapamycin (Wnt/mTOR) signaling, and altered lipid metabolism. Indeed, Wnt/mTOR activities were found to be modified by MPH in several ADHD studies. Wnt/mTOR was also found to play a role in sAD and in animal models of the disorder. Moreover, MPH treatment in the MCI phase was shown to be successful for apathy including some improvement in cognition, according to a recent meta-analysis. In several AD animal models, ADHD-like behavioral phenotypes have been observed indicating a possible interconnection between ADHD and AD. In this concept paper, we will discuss the various evidence in human and animal models supporting the hypothesis in which ADHD might increase the risk for sAD, with common involvement of the Wnt/mTOR-pathway leading to lifespan alteration at the neuronal levels.
Collapse
Affiliation(s)
- Edna Grünblatt
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich (PUK), University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and the Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Jan Homolak
- Department of Pharmacology and Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ana Babic Perhoc
- Department of Pharmacology and Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Virag Davor
- Department of Pharmacology and Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ana Knezovic
- Department of Pharmacology and Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Jelena Osmanovic Barilar
- Department of Pharmacology and Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Peter Riederer
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany.,Department and Research Unit of Psychiatry, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Susanne Walitza
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich (PUK), University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and the Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Christian Tackenberg
- Neuroscience Center Zurich, University of Zurich and the Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.,Institute for Regenerative Medicine (IREM), University of Zurich, Schlieren, Switzerland
| | - Melita Salkovic-Petrisic
- Department of Pharmacology and Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
11
|
Cannabinoid CB2 Receptors in Neurodegenerative Proteinopathies: New Insights and Therapeutic Potential. Biomedicines 2022; 10:biomedicines10123000. [PMID: 36551756 PMCID: PMC9775106 DOI: 10.3390/biomedicines10123000] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Some of the most prevalent neurodegenerative disorders, including Alzheimer's and Parkinson's disease, are proteinopathies characterized by the accumulation of specific protein aggregates in the brain. Such misfolded protein aggregates can trigger modulation of the innate and adaptive immune systems and subsequently lead to chronic neuroinflammation that drives the onset and progression of neurodegenerative diseases. Since there is still no effective disease-modifying treatment, new therapeutic targets for neurodegenerative proteinopathies have been sought. The endocannabinoid system, and in particular the cannabinoid CB2 receptors, have been extensively studied, due to their important role in neuroinflammation, especially in microglial cells. Several studies have shown promising effects of CB2 receptor activation on reducing protein aggregation-based pathology as well as on attenuating inflammation and several dementia-related symptoms. In this review, we discuss the available data on the role of CB2 receptors in neuroinflammation and the potential benefits and limitations of specific agonists of these receptors in the therapy of neurodegenerative proteinopathies.
Collapse
|
12
|
ENT-A010, a Novel Steroid Derivative, Displays Neuroprotective Functions and Modulates Microglial Responses. Biomolecules 2022; 12:biom12030424. [PMID: 35327616 PMCID: PMC8946810 DOI: 10.3390/biom12030424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/02/2022] [Accepted: 03/05/2022] [Indexed: 11/17/2022] Open
Abstract
Tackling neurodegeneration and neuroinflammation is particularly challenging due to the complexity of central nervous system (CNS) disorders, as well as the limited drug accessibility to the brain. The activation of tropomyosin-related kinase A (TRKA) receptor signaling by the nerve growth factor (NGF) or the neurosteroid dehydroepiandrosterone (DHEA) may combat neurodegeneration and regulate microglial function. In the present study, we synthesized a C-17-spiro-cyclopropyl DHEA derivative (ENT-A010), which was capable of activating TRKA. ENT-A010 protected PC12 cells against serum starvation-induced cell death, dorsal root ganglia (DRG) neurons against NGF deprivation-induced apoptosis and hippocampal neurons against Aβ-induced apoptosis. In addition, ENT-A010 pretreatment partially restored homeostatic features of microglia in the hippocampus of lipopolysaccharide (LPS)-treated mice, enhanced Aβ phagocytosis, and increased Ngf expression in microglia in vitro. In conclusion, the small molecule ENT-A010 elicited neuroprotective effects and modulated microglial function, thereby emerging as an interesting compound, which merits further study in the treatment of CNS disorders.
Collapse
|
13
|
Neurogenetic disorders across the lifespan: from aberrant development to degeneration. Nat Rev Neurol 2022; 18:117-124. [PMID: 34987232 PMCID: PMC10132523 DOI: 10.1038/s41582-021-00595-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2021] [Indexed: 02/08/2023]
Abstract
Intellectual disability and autism spectrum disorder (ASD) are common, and genetic testing is increasingly performed in individuals with these diagnoses to inform prognosis, refine management and provide information about recurrence risk in the family. For neurogenetic conditions associated with intellectual disability and ASD, data on natural history in adults are scarce; however, as older adults with these disorders are identified, it is becoming clear that some conditions are associated with both neurodevelopmental problems and neurodegeneration. Moreover, emerging evidence indicates that some neurogenetic conditions associated primarily with neurodegeneration also affect neurodevelopment. In this Perspective, we discuss examples of diseases that have developmental and degenerative overlap. We propose that neurogenetic disorders should be studied continually across the lifespan to understand the roles of the affected genes in brain development and maintenance, and to inform strategies for treatment.
Collapse
|
14
|
Schilder BM, Navarro E, Raj T. Multi-omic insights into Parkinson's Disease: From genetic associations to functional mechanisms. Neurobiol Dis 2021; 163:105580. [PMID: 34871738 PMCID: PMC10101343 DOI: 10.1016/j.nbd.2021.105580] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/17/2021] [Accepted: 12/02/2021] [Indexed: 02/07/2023] Open
Abstract
Genome-Wide Association Studies (GWAS) have elucidated the genetic components of Parkinson's Disease (PD). However, because the vast majority of GWAS association signals fall within non-coding regions, translating these results into an interpretable, mechanistic understanding of the disease etiology remains a major challenge in the field. In this review, we provide an overview of the approaches to prioritize putative causal variants and genes as well as summarise the primary findings of previous studies. We then discuss recent efforts to integrate multi-omics data to identify likely pathogenic cell types and biological pathways implicated in PD pathogenesis. We have compiled full summary statistics of cell-type, tissue, and phentoype enrichment analyses from multiple studies of PD GWAS and provided them in a standardized format as a resource for the research community (https://github.com/RajLabMSSM/PD_omics_review). Finally, we discuss the experimental, computational, and conceptual advances that will be necessary to fully elucidate the effects of functional variants and genes on cellular dysregulation and disease risk.
Collapse
Affiliation(s)
- Brian M Schilder
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; UK Dementia Research Institute at Imperial College London, London, United Kingdom.
| | - Elisa Navarro
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Sección Departamental de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Towfique Raj
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
15
|
Kisby GE, Spencer PS. Genotoxic Damage During Brain Development Presages Prototypical Neurodegenerative Disease. Front Neurosci 2021; 15:752153. [PMID: 34924930 PMCID: PMC8675606 DOI: 10.3389/fnins.2021.752153] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/20/2021] [Indexed: 01/15/2023] Open
Abstract
Western Pacific Amyotrophic Lateral Sclerosis and Parkinsonism-Dementia Complex (ALS/PDC) is a disappearing prototypical neurodegenerative disorder (tau-dominated polyproteinopathy) linked with prior exposure to phytogenotoxins in cycad seed used for medicine and/or food. The principal cycad genotoxin, methylazoxymethanol (MAM), forms reactive carbon-centered ions that alkylate nucleic acids in fetal rodent brain and, depending on the timing of systemic administration, induces persistent developmental abnormalities of the cortex, hippocampus, cerebellum, and retina. Whereas administration of MAM prenatally or postnatally can produce animal models of epilepsy, schizophrenia or ataxia, administration to adult animals produces little effect on brain structure or function. The neurotoxic effects of MAM administered to rats during cortical brain development (specifically, gestation day 17) are used to model the histological, neurophysiological and behavioral deficits of human schizophrenia, a condition that may precede or follow clinical onset of motor neuron disease in subjects with sporadic ALS and ALS/PDC. While studies of migrants to and from communities impacted by ALS/PDC indicate the degenerative brain disorder may be acquired in juvenile and adult life, a proportion of indigenous cases shows neurodevelopmental aberrations in the cerebellum and retina consistent with MAM exposure in utero. MAM induces specific patterns of DNA damage and repair that associate with increased tau expression in primary rat neuronal cultures and with brain transcriptional changes that parallel those associated with human ALS and Alzheimer's disease. We examine MAM in relation to neurodevelopment, epigenetic modification, DNA damage/replicative stress, genomic instability, somatic mutation, cell-cycle reentry and cellular senescence. Since the majority of neurodegenerative disease lacks a solely inherited genetic basis, research is needed to explore the hypothesis that early-life exposure to genotoxic agents may trigger or promote molecular events that culminate in neurodegeneration.
Collapse
Affiliation(s)
- Glen E. Kisby
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Lebanon, OR, United States
| | - Peter S. Spencer
- School of Medicine (Neurology), Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
16
|
Tsuchida A, Laurent A, Crivello F, Petit L, Joliot M, Pepe A, Beguedou N, Gueye MF, Verrecchia V, Nozais V, Zago L, Mellet E, Debette S, Tzourio C, Mazoyer B. The MRi-Share database: brain imaging in a cross-sectional cohort of 1870 university students. Brain Struct Funct 2021; 226:2057-2085. [PMID: 34283296 DOI: 10.1007/s00429-021-02334-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/11/2021] [Indexed: 01/04/2023]
Abstract
We report on MRi-Share, a multi-modal brain MRI database acquired in a unique sample of 1870 young healthy adults, aged 18-35 years, while undergoing university-level education. MRi-Share contains structural (T1 and FLAIR), diffusion (multispectral), susceptibility-weighted (SWI), and resting-state functional imaging modalities. Here, we described the contents of these different neuroimaging datasets and the processing pipelines used to derive brain phenotypes, as well as how quality control was assessed. In addition, we present preliminary results on associations of some of these brain image-derived phenotypes at the whole brain level with both age and sex, in the subsample of 1722 individuals aged less than 26 years. We demonstrate that the post-adolescence period is characterized by changes in both structural and microstructural brain phenotypes. Grey matter cortical thickness, surface area and volume were found to decrease with age, while white matter volume shows increase. Diffusivity, either radial or axial, was found to robustly decrease with age whereas fractional anisotropy only slightly increased. As for the neurite orientation dispersion and densities, both were found to increase with age. The isotropic volume fraction also showed a slight increase with age. These preliminary findings emphasize the complexity of changes in brain structure and function occurring in this critical period at the interface of late maturation and early ageing.
Collapse
Affiliation(s)
- Ami Tsuchida
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, Université de Bordeaux, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CNRS, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CEA, Bordeaux, France
| | - Alexandre Laurent
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, Université de Bordeaux, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CNRS, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CEA, Bordeaux, France
| | - Fabrice Crivello
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, Université de Bordeaux, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CNRS, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CEA, Bordeaux, France
| | - Laurent Petit
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, Université de Bordeaux, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CNRS, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CEA, Bordeaux, France
| | - Marc Joliot
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, Université de Bordeaux, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CNRS, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CEA, Bordeaux, France.,Ginesislab, Fealinx and Université de Bordeaux, Bordeaux, France
| | - Antonietta Pepe
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, Université de Bordeaux, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CNRS, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CEA, Bordeaux, France
| | - Naka Beguedou
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, Université de Bordeaux, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CNRS, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CEA, Bordeaux, France
| | - Marie-Fateye Gueye
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, Université de Bordeaux, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CNRS, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CEA, Bordeaux, France.,Ginesislab, Fealinx and Université de Bordeaux, Bordeaux, France
| | - Violaine Verrecchia
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, Université de Bordeaux, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CNRS, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CEA, Bordeaux, France.,Ginesislab, Fealinx and Université de Bordeaux, Bordeaux, France
| | - Victor Nozais
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, Université de Bordeaux, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CNRS, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CEA, Bordeaux, France.,Ginesislab, Fealinx and Université de Bordeaux, Bordeaux, France
| | - Laure Zago
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, Université de Bordeaux, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CNRS, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CEA, Bordeaux, France
| | - Emmanuel Mellet
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, Université de Bordeaux, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CNRS, Bordeaux, France.,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CEA, Bordeaux, France
| | - Stéphanie Debette
- Université de Bordeaux, INSERM, Bordeaux Population Health Research Center, U1219, CHU Bordeaux, Bordeaux, France.,Centre Hospitalier Universitaire Pellegrin, Bordeaux, France
| | - Christophe Tzourio
- Université de Bordeaux, INSERM, Bordeaux Population Health Research Center, U1219, CHU Bordeaux, Bordeaux, France.,Centre Hospitalier Universitaire Pellegrin, Bordeaux, France
| | - Bernard Mazoyer
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, Université de Bordeaux, Bordeaux, France. .,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CNRS, Bordeaux, France. .,Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR5293, CEA, Bordeaux, France. .,Ginesislab, Fealinx and Université de Bordeaux, Bordeaux, France. .,Centre Hospitalier Universitaire Pellegrin, Bordeaux, France.
| |
Collapse
|
17
|
Carbamate group as structural motif in drugs: a review of carbamate derivatives used as therapeutic agents. Arh Hig Rada Toksikol 2020; 71:285-299. [PMID: 33410773 PMCID: PMC7968508 DOI: 10.2478/aiht-2020-71-3466] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 12/01/2020] [Indexed: 12/17/2022] Open
Abstract
Due to their very good chemical and proteolytic stability, ability to penetrate cell membranes, and resemblance to a peptide bond, carbamate derivatives have received much attention in recent years and got an important role in modern drug discovery and medicinal chemistry. Today, carbamates make structural and/or functional part of many drugs and prodrugs approved and marketed for the treatment of various diseases such as cancer, epilepsy, hepatitis C, HIV infection, and Alzheimer's disease. In drugs they can play a role in drug-target interaction or improve the biological activity of parent molecules. In prodrugs they are mainly used to delay first-pass metabolism and enhance the bioavailability and effectiveness of compounds. This brief review takes a look at the properties and use of carbamates in various fields of medicine and provides quick insights into the mechanisms of action for some of them.
Collapse
|
18
|
Gagliardi D, Costamagna G, Taiana M, Andreoli L, Biella F, Bersani M, Bresolin N, Comi GP, Corti S. Insights into disease mechanisms and potential therapeutics for C9orf72-related amyotrophic lateral sclerosis/frontotemporal dementia. Ageing Res Rev 2020; 64:101172. [PMID: 32971256 DOI: 10.1016/j.arr.2020.101172] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022]
Abstract
In 2011, a hexanucleotide repeat expansion (HRE) in the noncoding region of C9orf72 was associated with the most frequent genetic cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). The main pathogenic mechanisms in C9-ALS/FTD are haploinsufficiency of the C9orf72 protein and gain of function toxicity from bidirectionally-transcribed repeat-containing RNAs and dipeptide repeat proteins (DPRs) resulting from non-canonical RNA translation. Additionally, abnormalities in different downstream cellular mechanisms, such as nucleocytoplasmic transport and autophagy, play a role in pathogenesis. Substantial research efforts using in vitro and in vivo models have provided valuable insights into the contribution of each mechanism in disease pathogenesis. However, conflicting evidence exists, and a unifying theory still lacks. Here, we provide an overview of the recently published literature on clinical, neuropathological and molecular features of C9-ALS/FTD. We highlight the supposed neuronal role of C9orf72 and the HRE pathogenic cascade, mainly focusing on the contribution of RNA foci and DPRs to neurodegeneration and discussing the several downstream mechanisms. We summarize the emerging biochemical and neuroimaging biomarkers, as well as the potential therapeutic approaches. Despite promising results, a specific disease-modifying treatment is still not available to date and greater insights into disease mechanisms may help in this direction.
Collapse
Affiliation(s)
- Delia Gagliardi
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Gianluca Costamagna
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Michela Taiana
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Luca Andreoli
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Fabio Biella
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Margherita Bersani
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Nereo Bresolin
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy; Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Giacomo Pietro Comi
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy; Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy; Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy.
| |
Collapse
|
19
|
van Rooij J, Mol MO, Melhem S, van der Wal P, Arp P, Paron F, Donker Kaat L, Seelaar H, Netherlands Brain Bank, Miedema SSM, Oshima T, Eggen BJL, Uitterlinden A, van Meurs J, van Kesteren RE, Smit AB, Buratti E, van Swieten JC. Somatic TARDBP variants as a cause of semantic dementia. Brain 2020; 143:3827-3841. [PMID: 33155043 PMCID: PMC7805802 DOI: 10.1093/brain/awaa317] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/13/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
The aetiology of late-onset neurodegenerative diseases is largely unknown. Here we investigated whether de novo somatic variants for semantic dementia can be detected, thereby arguing for a more general role of somatic variants in neurodegenerative disease. Semantic dementia is characterized by a non-familial occurrence, early onset (<65 years), focal temporal atrophy and TDP-43 pathology. To test whether somatic variants in neural progenitor cells during brain development might lead to semantic dementia, we compared deep exome sequencing data of DNA derived from brain and blood of 16 semantic dementia cases. Somatic variants observed in brain tissue and absent in blood were validated using amplicon sequencing and digital PCR. We identified two variants in exon one of the TARDBP gene (L41F and R42H) at low level (1-3%) in cortical regions and in dentate gyrus in two semantic dementia brains, respectively. The pathogenicity of both variants is supported by demonstrating impaired splicing regulation of TDP-43 and by altered subcellular localization of the mutant TDP-43 protein. These findings indicate that somatic variants may cause semantic dementia as a non-hereditary neurodegenerative disease, which might be exemplary for other late-onset neurodegenerative disorders.
Collapse
Affiliation(s)
- Jeroen van Rooij
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Merel O Mol
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Shamiram Melhem
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Pelle van der Wal
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Pascal Arp
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Francesca Paron
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Laura Donker Kaat
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Harro Seelaar
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Suzanne S M Miedema
- Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, The Netherlands
| | - Takuya Oshima
- Department of Biomedical Sciences of Cells and Systems, section Molecular Neurobiology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Bart J L Eggen
- Department of Biomedical Sciences of Cells and Systems, section Molecular Neurobiology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - André Uitterlinden
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Joyce van Meurs
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ronald E van Kesteren
- Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, The Netherlands
| | - August B Smit
- Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, The Netherlands
| | - Emanuele Buratti
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - John C van Swieten
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
20
|
Sánchez-Torres JL, Yescas-Gómez P, Torres-Romero J, Espinosa OR, Canovas LL, Tecalco-Cruz ÁC, Ponce-Regalado MD, Alvarez-Sánchez ME. Matrix metalloproteinases deregulation in amyotrophic lateral sclerosis. J Neurol Sci 2020; 419:117175. [PMID: 33068904 DOI: 10.1016/j.jns.2020.117175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/05/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the loss of upper and lower motor neurons that results in progressive paralysis and muscular atrophy. There are many molecules and genes involved in neuromuscular degeneration in ALS; among these, matrix metalloproteinases (MMPs). MMPs play an important role in the pathology of ALS, and MMP-1, 2, 3, and 9 might serve as disease progression markers. Tissue inhibitors of metalloproteinases (TIMPS) might also function as progression markers in ALS because they participate in regulating the proteolytic activity of MMPs. Moreover, a diversity of genes also plays a role in the pathogenesis of ALS; most MMPs-coding genes present variants related to the pathological proteolytic activity. This short review, however, will focus on the role of matrix metalloproteinases in ALS.
Collapse
Affiliation(s)
- Jorge Luis Sánchez-Torres
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México (UACM), San Lorenzo 290, Del Valle Sur, Benito Juárez, 03100 Mexico City, Mexico
| | - Petra Yescas-Gómez
- Department of Genetics, National Institute of Neurology and Neurosurgery "Manuel Velasco Suárez", . Insurgentes Sur 3877 La Fama, Tlalpan, 14269 Mexico City, Mexico
| | - Julio Torres-Romero
- Facultad de Química, Universidad Autónoma de Yucatán (UADY), Calle 43 S/N entre calle 96 y calle 40, Colonia Inalámbrica, C.P. 97069, Mérida, Yucatán, Mexico
| | - Oscar Rojas Espinosa
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Carpio y Plan de Ayala, Colonia Santo Tomás, 11340, Mexico City, Mexico
| | - Lilia López Canovas
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México (UACM), San Lorenzo 290, Del Valle Sur, Benito Juárez, 03100 Mexico City, Mexico
| | - Ángeles C Tecalco-Cruz
- Bachelor of Science in Genomics, Autonomous University of Mexico City (UACM). San Lorenzo 290, Del Valle Sur, Benito Juárez, 03100 Mexico City, Mexico
| | - María Dolores Ponce-Regalado
- Departamento de Ciencias de la Salud, Centro Universitario de los Altos, Universidad de Guadalajara Av. Rafael Casillas Aceves No. 1200, Tepatitlán, de Morelos, 47610, Jalisco, Mexico
| | - María Elizbeth Alvarez-Sánchez
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México (UACM), San Lorenzo 290, Del Valle Sur, Benito Juárez, 03100 Mexico City, Mexico.
| |
Collapse
|
21
|
Walhovd KB, Fjell AM, Sørensen Ø, Mowinckel AM, Reinbold CS, Idland AV, Watne LO, Franke A, Dobricic V, Kilpert F, Bertram L, Wang Y. Genetic risk for Alzheimer disease predicts hippocampal volume through the human lifespan. NEUROLOGY-GENETICS 2020; 6:e506. [PMID: 33134508 PMCID: PMC7577559 DOI: 10.1212/nxg.0000000000000506] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 07/17/2020] [Indexed: 11/27/2022]
Abstract
Objective To test the hypothesis that genetic risk for Alzheimer disease (AD) may represent a stable influence on the brain from early in life, rather than being primarily age dependent, we investigated in a lifespan sample of 1,181 persons with a total of 2,690 brain scans, whether higher polygenic risk score (PGS) for AD and presence of APOE ε4 was associated with lower hippocampal volumes to begin with, as an offset effect, or possibly faster decline in older age. Methods Using general additive mixed models, we assessed the relations of PGS for AD, including variants in APOE with hippocampal volume and its change in a cognitively healthy longitudinal lifespan sample (age range: 4–95 years, mean visit age 39.7 years, SD 26.9 years), followed for up to 11 years. Results AD-PGS and APOE ε4 in isolation showed a significant negative effect on hippocampal volume. The effect of a 1 sample SD increase in AD-PGS on hippocampal volume was estimated to –36.4 mm3 (confidence interval [CI]: –71.8, –1.04) and the effect of carrying ε4 allele(s) –107.0 mm3 (CI: –182.0, –31.5). Offset effects of AD-PGS and APOE ε4 were present in hippocampal development, and interactions between age and genetic risk on volume change were not consistently observed. Conclusions Endophenotypic manifestation of polygenic risk for AD may be seen across the lifespan in cognitively healthy persons, not being confined to clinical populations or older age. This emphasizes that a broader population and age range may be relevant targets for attempts to prevent AD.
Collapse
Affiliation(s)
- Kristine B Walhovd
- Center for Lifespan Changes in Brain and Cognition (K.B.W., A.M.F., Ø.S., A.M.M., C.S.R., A.-V.I., L.B., Y.W.), Department of Psychology, University of Oslo; Division of Radiology and Nuclear Medicine (K.B.W., A.M.F.), Oslo University Hospital, Rikshospitalet; Oslo Delirium Research Group (A.-V.I., L.O.W.), Department of Geriatric Medicine, and Institute of Basic Medical Sciences (A.-V.I., L.O.W.), University of Oslo, Norway; Institute of Clinical Molecular Biology (A.F.), Christian-Albrechts-University of Kiel; and Lübeck Interdisciplinary Platform for Genome Analytics (V.D., F.K., L.B.), Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Germany
| | - Anders M Fjell
- Center for Lifespan Changes in Brain and Cognition (K.B.W., A.M.F., Ø.S., A.M.M., C.S.R., A.-V.I., L.B., Y.W.), Department of Psychology, University of Oslo; Division of Radiology and Nuclear Medicine (K.B.W., A.M.F.), Oslo University Hospital, Rikshospitalet; Oslo Delirium Research Group (A.-V.I., L.O.W.), Department of Geriatric Medicine, and Institute of Basic Medical Sciences (A.-V.I., L.O.W.), University of Oslo, Norway; Institute of Clinical Molecular Biology (A.F.), Christian-Albrechts-University of Kiel; and Lübeck Interdisciplinary Platform for Genome Analytics (V.D., F.K., L.B.), Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Germany
| | - Øystein Sørensen
- Center for Lifespan Changes in Brain and Cognition (K.B.W., A.M.F., Ø.S., A.M.M., C.S.R., A.-V.I., L.B., Y.W.), Department of Psychology, University of Oslo; Division of Radiology and Nuclear Medicine (K.B.W., A.M.F.), Oslo University Hospital, Rikshospitalet; Oslo Delirium Research Group (A.-V.I., L.O.W.), Department of Geriatric Medicine, and Institute of Basic Medical Sciences (A.-V.I., L.O.W.), University of Oslo, Norway; Institute of Clinical Molecular Biology (A.F.), Christian-Albrechts-University of Kiel; and Lübeck Interdisciplinary Platform for Genome Analytics (V.D., F.K., L.B.), Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Germany
| | - Athanasia Monika Mowinckel
- Center for Lifespan Changes in Brain and Cognition (K.B.W., A.M.F., Ø.S., A.M.M., C.S.R., A.-V.I., L.B., Y.W.), Department of Psychology, University of Oslo; Division of Radiology and Nuclear Medicine (K.B.W., A.M.F.), Oslo University Hospital, Rikshospitalet; Oslo Delirium Research Group (A.-V.I., L.O.W.), Department of Geriatric Medicine, and Institute of Basic Medical Sciences (A.-V.I., L.O.W.), University of Oslo, Norway; Institute of Clinical Molecular Biology (A.F.), Christian-Albrechts-University of Kiel; and Lübeck Interdisciplinary Platform for Genome Analytics (V.D., F.K., L.B.), Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Germany
| | - Céline Sonja Reinbold
- Center for Lifespan Changes in Brain and Cognition (K.B.W., A.M.F., Ø.S., A.M.M., C.S.R., A.-V.I., L.B., Y.W.), Department of Psychology, University of Oslo; Division of Radiology and Nuclear Medicine (K.B.W., A.M.F.), Oslo University Hospital, Rikshospitalet; Oslo Delirium Research Group (A.-V.I., L.O.W.), Department of Geriatric Medicine, and Institute of Basic Medical Sciences (A.-V.I., L.O.W.), University of Oslo, Norway; Institute of Clinical Molecular Biology (A.F.), Christian-Albrechts-University of Kiel; and Lübeck Interdisciplinary Platform for Genome Analytics (V.D., F.K., L.B.), Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Germany
| | - Ane-Victoria Idland
- Center for Lifespan Changes in Brain and Cognition (K.B.W., A.M.F., Ø.S., A.M.M., C.S.R., A.-V.I., L.B., Y.W.), Department of Psychology, University of Oslo; Division of Radiology and Nuclear Medicine (K.B.W., A.M.F.), Oslo University Hospital, Rikshospitalet; Oslo Delirium Research Group (A.-V.I., L.O.W.), Department of Geriatric Medicine, and Institute of Basic Medical Sciences (A.-V.I., L.O.W.), University of Oslo, Norway; Institute of Clinical Molecular Biology (A.F.), Christian-Albrechts-University of Kiel; and Lübeck Interdisciplinary Platform for Genome Analytics (V.D., F.K., L.B.), Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Germany
| | - Leiv Otto Watne
- Center for Lifespan Changes in Brain and Cognition (K.B.W., A.M.F., Ø.S., A.M.M., C.S.R., A.-V.I., L.B., Y.W.), Department of Psychology, University of Oslo; Division of Radiology and Nuclear Medicine (K.B.W., A.M.F.), Oslo University Hospital, Rikshospitalet; Oslo Delirium Research Group (A.-V.I., L.O.W.), Department of Geriatric Medicine, and Institute of Basic Medical Sciences (A.-V.I., L.O.W.), University of Oslo, Norway; Institute of Clinical Molecular Biology (A.F.), Christian-Albrechts-University of Kiel; and Lübeck Interdisciplinary Platform for Genome Analytics (V.D., F.K., L.B.), Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Germany
| | - Andre Franke
- Center for Lifespan Changes in Brain and Cognition (K.B.W., A.M.F., Ø.S., A.M.M., C.S.R., A.-V.I., L.B., Y.W.), Department of Psychology, University of Oslo; Division of Radiology and Nuclear Medicine (K.B.W., A.M.F.), Oslo University Hospital, Rikshospitalet; Oslo Delirium Research Group (A.-V.I., L.O.W.), Department of Geriatric Medicine, and Institute of Basic Medical Sciences (A.-V.I., L.O.W.), University of Oslo, Norway; Institute of Clinical Molecular Biology (A.F.), Christian-Albrechts-University of Kiel; and Lübeck Interdisciplinary Platform for Genome Analytics (V.D., F.K., L.B.), Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Germany
| | - Valerija Dobricic
- Center for Lifespan Changes in Brain and Cognition (K.B.W., A.M.F., Ø.S., A.M.M., C.S.R., A.-V.I., L.B., Y.W.), Department of Psychology, University of Oslo; Division of Radiology and Nuclear Medicine (K.B.W., A.M.F.), Oslo University Hospital, Rikshospitalet; Oslo Delirium Research Group (A.-V.I., L.O.W.), Department of Geriatric Medicine, and Institute of Basic Medical Sciences (A.-V.I., L.O.W.), University of Oslo, Norway; Institute of Clinical Molecular Biology (A.F.), Christian-Albrechts-University of Kiel; and Lübeck Interdisciplinary Platform for Genome Analytics (V.D., F.K., L.B.), Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Germany
| | - Fabian Kilpert
- Center for Lifespan Changes in Brain and Cognition (K.B.W., A.M.F., Ø.S., A.M.M., C.S.R., A.-V.I., L.B., Y.W.), Department of Psychology, University of Oslo; Division of Radiology and Nuclear Medicine (K.B.W., A.M.F.), Oslo University Hospital, Rikshospitalet; Oslo Delirium Research Group (A.-V.I., L.O.W.), Department of Geriatric Medicine, and Institute of Basic Medical Sciences (A.-V.I., L.O.W.), University of Oslo, Norway; Institute of Clinical Molecular Biology (A.F.), Christian-Albrechts-University of Kiel; and Lübeck Interdisciplinary Platform for Genome Analytics (V.D., F.K., L.B.), Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Germany
| | - Lars Bertram
- Center for Lifespan Changes in Brain and Cognition (K.B.W., A.M.F., Ø.S., A.M.M., C.S.R., A.-V.I., L.B., Y.W.), Department of Psychology, University of Oslo; Division of Radiology and Nuclear Medicine (K.B.W., A.M.F.), Oslo University Hospital, Rikshospitalet; Oslo Delirium Research Group (A.-V.I., L.O.W.), Department of Geriatric Medicine, and Institute of Basic Medical Sciences (A.-V.I., L.O.W.), University of Oslo, Norway; Institute of Clinical Molecular Biology (A.F.), Christian-Albrechts-University of Kiel; and Lübeck Interdisciplinary Platform for Genome Analytics (V.D., F.K., L.B.), Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Germany
| | - Yunpeng Wang
- Center for Lifespan Changes in Brain and Cognition (K.B.W., A.M.F., Ø.S., A.M.M., C.S.R., A.-V.I., L.B., Y.W.), Department of Psychology, University of Oslo; Division of Radiology and Nuclear Medicine (K.B.W., A.M.F.), Oslo University Hospital, Rikshospitalet; Oslo Delirium Research Group (A.-V.I., L.O.W.), Department of Geriatric Medicine, and Institute of Basic Medical Sciences (A.-V.I., L.O.W.), University of Oslo, Norway; Institute of Clinical Molecular Biology (A.F.), Christian-Albrechts-University of Kiel; and Lübeck Interdisciplinary Platform for Genome Analytics (V.D., F.K., L.B.), Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Germany
| |
Collapse
|
22
|
Rajpoot K. Nanotechnology-based Targeting of Neurodegenerative Disorders: A Promising Tool for Efficient Delivery of Neuromedicines. Curr Drug Targets 2020; 21:819-836. [PMID: 31906836 DOI: 10.2174/1389450121666200106105633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/18/2019] [Accepted: 12/18/2019] [Indexed: 12/13/2022]
Abstract
Traditional drug delivery approaches remained ineffective in offering better treatment to various neurodegenerative disorders (NDs). In this context, diverse types of nanocarriers have shown their great potential to cross the blood-brain barrier (BBB) and have emerged as a prominent carrier system in drug delivery. Moreover, nanotechnology-based methods usually involve numerous nanosized carrier platforms, which potentiate the effect of the therapeutic agents in the therapy of NDs especially in diagnosis and drug delivery with negligible side effects. In addition, nanotechnology-based techniques have offered several strategies to cross BBB to intensify the bioavailability of drug moieties in the brain. In the last few years, diverse kinds of nanoparticles (NPs) have been developed by incorporating various biocompatible components (e.g., polysaccharide-based NPs, polymeric NPs, selenium NPs, AuNPs, protein-based NPs, gadolinium NPs, etc.), that showed great therapeutic benefits against NDs. Eventually, this review provides deep insights to explore recent applications of some innovative nanocarriers enclosing active molecules for the efficient treatment of NDs.
Collapse
Affiliation(s)
- Kuldeep Rajpoot
- Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, 495 009, Chhattisgarh, India
| |
Collapse
|
23
|
The puzzle of preserved cognition in the oldest old. Neurol Sci 2019; 41:441-447. [PMID: 31713754 DOI: 10.1007/s10072-019-04111-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 10/15/2019] [Indexed: 02/07/2023]
Abstract
Although epidemiological studies predict an exponential increase in the prevalence of dementia with age, recent studies have demonstrated that the oldest old are actually less frequently affected by dementia than the younger elderly. To explain this, I suggest a parallel between brain ageing and Alzheimer's disease (AD) and assume that theories concerning the brain's vulnerability to AD and its individual variability may also explain why some of the oldest old remain cognitively efficient. Some theories argue that AD is due to the continuing presence of the immature neurones vulnerable to amyloid beta protein (Aß) that are normally involved in brain development and then removed as a result of cell selection by the proteins associated with both brain development and AD. If a dysfunction in cell selection allows these immature neurones to survive, they degenerate early as a result of the neurotoxic action of Aß accumulation, which their mature counterparts can withstand. Consequently, age at the time of onset of AD and its clinical presentations depend on the number and location of such immature cells. I speculate that the same mechanism is responsible for the variability of normal brain ageing: the oldest old with well-preserved cognitive function are people genetically programmed for extreme ageing who have benefited from better cell selection during prenatal and neonatal life and therefore have fewer surviving neurones vulnerable to amyloid-promoted degeneration, whereas the process of early life cell selection was less successful in the oldest old who develop dementia.
Collapse
|
24
|
Raghu P, Joseph A, Krishnan H, Singh P, Saha S. Phosphoinositides: Regulators of Nervous System Function in Health and Disease. Front Mol Neurosci 2019; 12:208. [PMID: 31507376 PMCID: PMC6716428 DOI: 10.3389/fnmol.2019.00208] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 08/07/2019] [Indexed: 12/11/2022] Open
Abstract
Phosphoinositides, the seven phosphorylated derivatives of phosphatidylinositol have emerged as regulators of key sub-cellular processes such as membrane transport, cytoskeletal function and plasma membrane signaling in eukaryotic cells. All of these processes are also present in the cells that constitute the nervous system of animals and in this setting too, these are likely to tune key aspects of cell biology in relation to the unique structure and function of neurons. Phosphoinositides metabolism and function are mediated by enzymes and proteins that are conserved in evolution, and analysis of knockouts of these in animal models implicate this signaling system in neural function. Most recently, with the advent of human genome analysis, mutations in genes encoding components of the phosphoinositide signaling pathway have been implicated in human diseases although the cell biological basis of disease phenotypes in many cases remains unclear. In this review we evaluate existing evidence for the involvement of phosphoinositide signaling in human nervous system diseases and discuss ways of enhancing our understanding of the role of this pathway in the human nervous system's function in health and disease.
Collapse
Affiliation(s)
- Padinjat Raghu
- National Centre for Biological Sciences-TIFR, Bengaluru, India
| | | | | | | | | |
Collapse
|
25
|
Penke B, Bogár F, Paragi G, Gera J, Fülöp L. Key Peptides and Proteins in Alzheimer's Disease. Curr Protein Pept Sci 2019; 20:577-599. [PMID: 30605056 DOI: 10.2174/1389203720666190103123434] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/03/2018] [Accepted: 12/27/2018] [Indexed: 02/02/2023]
Abstract
Alzheimer's Disease (AD) is a form of progressive dementia involving cognitive impairment, loss of learning and memory. Different proteins (such as amyloid precursor protein (APP), β- amyloid (Aβ) and tau protein) play a key role in the initiation and progression of AD. We review the role of the most important proteins and peptides in AD pathogenesis. The structure, biosynthesis and physiological role of APP are shortly summarized. The details of trafficking and processing of APP to Aβ, the cytosolic intracellular Aβ domain (AICD) and small soluble proteins are shown, together with other amyloid-forming proteins such as tau and α-synuclein (α-syn). Hypothetic physiological functions of Aβ are summarized. The mechanism of conformational change, the formation and the role of neurotoxic amyloid oligomeric (oAβ) are shown. The fibril formation process and the co-existence of different steric structures (U-shaped and S-shaped) of Aβ monomers in mature fibrils are demonstrated. We summarize the known pathogenic and non-pathogenic mutations and show the toxic interactions of Aβ species after binding to cellular receptors. Tau phosphorylation, fibrillation, the molecular structure of tau filaments and their toxic effect on microtubules are shown. Development of Aβ and tau imaging in AD brain and CSF as well as blood biomarkers is shortly summarized. The most probable pathomechanisms of AD including the toxic effects of oAβ and tau; the three (biochemical, cellular and clinical) phases of AD are shown. Finally, the last section summarizes the present state of Aβ- and tau-directed therapies and future directions of AD research and drug development.
Collapse
Affiliation(s)
- Botond Penke
- Department of Medical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dom square 8, Szeged, H-6720, Hungary
| | - Ferenc Bogár
- Department of Medical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dom square 8, Szeged, H-6720, Hungary.,MTA-SZTE Biomimetic Systems Research Group, University of Szeged, H-6720 Szeged, Dom square 8, Hungary
| | - Gábor Paragi
- MTA-SZTE Biomimetic Systems Research Group, University of Szeged, H-6720 Szeged, Dom square 8, Hungary.,Institute of Physics, University of Pécs, H-7624 Pecs, Ifjusag utja 6, Hungary
| | - János Gera
- Department of Medical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dom square 8, Szeged, H-6720, Hungary
| | - Lívia Fülöp
- Department of Medical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dom square 8, Szeged, H-6720, Hungary
| |
Collapse
|
26
|
Abstract
Millions of Americans now entering midlife and old age were exposed to high levels of lead, a neurotoxin, as children. Evidence from animal-model and human observational studies suggest that childhood lead exposure may raise the risk of adult neurodegenerative disease, particularly dementia, through a variety of possible mechanisms including epigenetic modification, delayed cardiovascular and kidney disease, direct degenerative CNS injury from lead remobilized from bone, and lowered neural and cognitive reserve. Within the next ten years, the generation of children with the highest historical lead exposures, those born in the 1960s, 1970s, and 1980s, will begin to enter the age at which dementia symptoms tend to emerge. Many will also enter the age in which lead stored in the skeleton may be remobilized at greater rates, particularly for women entering menopause and men and women experiencing osteoporosis. Should childhood lead exposure prove pro-degenerative, the next twenty years will provide the last opportunities for possible early intervention to forestall greater degenerative disease burden across the aging lead-exposed population. More evidence is needed now to characterize the nature and magnitude of the degenerative risks facing adults exposed to lead as children and to identify interventions to limit long-term harm.
Collapse
Affiliation(s)
- Aaron Reuben
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| |
Collapse
|
27
|
de Wit NM, den Hoedt S, Martinez-Martinez P, Rozemuller AJ, Mulder MT, de Vries HE. Astrocytic ceramide as possible indicator of neuroinflammation. J Neuroinflammation 2019; 16:48. [PMID: 30803453 PMCID: PMC6388480 DOI: 10.1186/s12974-019-1436-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 02/13/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease dementia (PDD), and frontotemporal lobar dementia (FTLD) are characterized by progressive neuronal loss but differ in their underlying pathological mechanisms. However, neuroinflammation is commonly observed within these different forms of dementia. Recently, it has been suggested that an altered sphingolipid metabolism may contribute to the pathogenesis of a variety of neurodegenerative conditions. Especially ceramide, the precursor of all complex sphingolipids, is thought to be associated with pro-apoptotic cellular processes, thereby propagating neurodegeneration and neuroinflammation, although it remains unclear to what extent. The current pathological study therefore investigates whether increased levels of ceramide are associated with the degree of neuroinflammation in various neurodegenerative disorders. METHODS Immunohistochemistry was performed on human post-mortem tissue of PDD and FTLD Pick's disease cases, which are well-characterized cases of dementia subtypes differing in their neuroinflammatory status, to assess the expression and localization of ceramide, acid sphingomyelinase, and ceramide synthase 2 and 5. In addition, we determined the concentration of sphingosine, sphingosine-1-phosphate (S1P), and ceramide species differing in their chain-length in brain homogenates of the post-mortem tissue using HPLC-MS/MS. RESULTS Our immunohistochemical analysis reveals that neuroinflammation is associated with increased ceramide levels in astrocytes in FTLD Pick's disease. Moreover, the observed increase in ceramide in astrocytes correlates with the expression of ceramide synthase 5. In addition, HPLC-MS/MS analysis shows a shift in ceramide species under neuroinflammatory conditions, favoring pro-apoptotic ceramide. CONCLUSIONS Together, these findings suggest that detected increased levels of pro-apoptotic ceramide might be a common denominator of neuroinflammation in different neurodegenerative diseases.
Collapse
Affiliation(s)
- Nienke M. de Wit
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands
| | - Sandra den Hoedt
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Pilar Martinez-Martinez
- Department of Neuroscience, School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Annemieke J. Rozemuller
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Monique T. Mulder
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Helga E. de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands
| |
Collapse
|
28
|
Dwivedi N, Shah J, Mishra V, Tambuwala M, Kesharwani P. Nanoneuromedicine for management of neurodegenerative disorder. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2018.12.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
29
|
Reid G. Disentangling What We Know About Microbes and Mental Health. Front Endocrinol (Lausanne) 2019; 10:81. [PMID: 30828318 PMCID: PMC6384226 DOI: 10.3389/fendo.2019.00081] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 01/30/2019] [Indexed: 12/14/2022] Open
Abstract
Much has been written in recent years about the gut-brain axis. Exciting pilot studies suggest probiotic applications to the gut can reduce anxiety and depression via the vagus nerve. But not to diminish such findings, much still needs to be considered, including the fact that the vagus nerve links to many other body sites that also host a microbiome. Questions remain that touch the core of being human: (i) Do our microbes influence happiness and to what extent? (ii) What components of the gut microbiota and their function, including as it relates to mental health, are critical and how do they differ between agile, fit hunter gatherers and obese westerners or Danes described as the happiest people on the planet? (iii) What role do environmental pollutants play in this microbes-host ecosystem? While approaching life from a reductionist perspective has a long history in science, we need to try to interrogate these health and disease issues from a wider perspective. For verification of a link between the gut microbiota and brain, and to test new therapies, human studies are needed, and are long overdue.
Collapse
Affiliation(s)
- Gregor Reid
- Canadian R&D Centre for Human Microbiome and Probiotics, Lawson Health Research Institute, London, ON, Canada
- Department of Microbiology and Immunology, and Surgery, Western University, London, ON, Canada
- *Correspondence: Gregor Reid
| |
Collapse
|
30
|
Aymerich MS, Aso E, Abellanas MA, Tolon RM, Ramos JA, Ferrer I, Romero J, Fernández-Ruiz J. Cannabinoid pharmacology/therapeutics in chronic degenerative disorders affecting the central nervous system. Biochem Pharmacol 2018; 157:67-84. [PMID: 30121249 DOI: 10.1016/j.bcp.2018.08.016] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/13/2018] [Indexed: 12/12/2022]
Abstract
The endocannabinoid system (ECS) exerts a modulatory effect of important functions such as neurotransmission, glial activation, oxidative stress, or protein homeostasis. Dysregulation of these cellular processes is a common neuropathological hallmark in aging and in neurodegenerative diseases of the central nervous system (CNS). The broad spectrum of actions of cannabinoids allows targeting different aspects of these multifactorial diseases. In this review, we examine the therapeutic potential of the ECS for the treatment of chronic neurodegenerative diseases of the CNS focusing on Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. First, we describe the localization of the molecular components of the ECS and how they are altered under neurodegenerative conditions, either contributing to or protecting cells from degeneration. Second, we address recent advances in the modulation of the ECS using experimental models through different strategies including the direct targeting of cannabinoid receptors with agonists or antagonists, increasing the endocannabinoid tone by the inhibition of endocannabinoid hydrolysis, and activation of cannabinoid receptor-independent effects. Preclinical evidence indicates that cannabinoid pharmacology is complex but supports the therapeutic potential of targeting the ECS. Third, we review the clinical evidence and discuss the future perspectives on how to bridge human and animal studies to develop cannabinoid-based therapies for each neurodegenerative disorder. Finally, we summarize the most relevant opportunities of cannabinoid pharmacology related to each disease and the multiple unexplored pathways in cannabinoid pharmacology that could be useful for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Maria S Aymerich
- Universidad de Navarra, Facultad de Ciencias, Departamento de Bioquímica y Genética, Pamplona, Spain; Universidad de Navarra, CIMA, Programa de Neurociencias, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Spain.
| | - Ester Aso
- Departamento de Patología y Terapéutica Experimental, Universidad de Barcelona, L'Hospitalet de Llobregat, Spain; CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Spain
| | - Miguel A Abellanas
- Universidad de Navarra, Facultad de Ciencias, Departamento de Bioquímica y Genética, Pamplona, Spain; Universidad de Navarra, CIMA, Programa de Neurociencias, Pamplona, Spain
| | - Rosa M Tolon
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Spain
| | - Jose A Ramos
- CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Spain; Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; IRYCIS, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Isidre Ferrer
- Departamento de Patología y Terapéutica Experimental, Universidad de Barcelona, L'Hospitalet de Llobregat, Spain; CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Spain
| | - Julian Romero
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Spain
| | - Javier Fernández-Ruiz
- CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Spain; Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; IRYCIS, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| |
Collapse
|
31
|
Yeh TH, Liu HF, Li YW, Lu CS, Shih HY, Chiu CC, Lin SJ, Huang YC, Cheng YC. C9orf72 is essential for neurodevelopment and motility mediated by Cyclin G1. Exp Neurol 2018. [PMID: 29522758 DOI: 10.1016/j.expneurol.2018.03.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hexanucleotide repeat expansions in the C9orf72 gene are a common genetic cause of familial and sporadic amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). However, the function of C9orf72 in neural development and the pathogenic mechanism underlying neurodegeneration are unknown. We found that disrupting C9orf72 expression by using C9orf72 constructs that lack the complete DENN domain result in reduced GTPase activity in zebrafish embryos, demonstrating the indispensability of the complete DENN domain. This effect was phenocopied by knocking down endogenous C9orf72 expression by using morpholinos. C9orf72-deficient zebrafish embryos exhibited impaired axonogenesis and motility defects. The C9orf72 deficiency upregulated the expression of tp53 and caused neuronal apoptosis. Knockdown Tp53 in the C9orf72-deficient embryos rescued only the apoptotic phenotype but not the phenotype with axonal and motility defects. The C9orf72 deficiency also induced ccng1 (encodes Cyclin G1) mRNA expression, and injection of a dominant-negative Cyclin G1 construct rescued the axonal impairment, apoptosis, and motility defects in the C9orf72-deficient embryos. Our results revealed the GTPase activity of C9orf72 and demonstrated that Cyclin G1 is an essential downstream mediator for C9orf72 in neural development and motility. Furthermore, downregulating Cyclin G1 was sufficient to rescue all the defects caused by C9orf72 deficiency. In summary, we revealed a novel regulatory mechanism underlying the role of C9orf72 in neurological and motility defects. This result facilitates understanding the function of the C9orf72 gene in the developing nervous system and provides a potential mechanism underlying the pathogenesis of ALS-FTD.
Collapse
Affiliation(s)
- Tu-Hsueh Yeh
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan; Section of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou Medical Center, Taoyuan, Taiwan; Department of Neurology, Taipei Medical University Hospital, Taipei, Taiwan; School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Han-Fang Liu
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Wen Li
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chin-Song Lu
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan; Section of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou Medical Center, Taoyuan, Taiwan
| | - Hung-Yu Shih
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ching-Chi Chiu
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Sheng-Jia Lin
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yin-Cheng Huang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou Medical Center, Taoyuan, Taiwan.
| | - Yi-Chuan Cheng
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
32
|
Yu J, Zhu H, Perry S, Taheri S, Kindy MS. Daily supplementation with GrandFusion ® improves memory and learning in aged rats. Aging (Albany NY) 2017; 9:1041-1054. [PMID: 28351996 PMCID: PMC5391217 DOI: 10.18632/aging.101209] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 03/17/2017] [Indexed: 01/10/2023]
Abstract
Studies have shown that supplementation with extracts from various sources, including fruits and vegetables reverse the age-related changes in movement and cognition. We hypothesized that these beneficial effects result from the presence of anti-oxidants and anti-inflammatory compounds in the fruits and vegetables that contribute to reduced oxidative stress, inflammation and cell death while potentially enhancing neurogenesis. The present study was performed to determine the impact of supplementation with GrandFusion®(GF) to aged Fisher 344 rats for 4 months to determine the impact on attenuation or reversal of the age-related deficits. When the aged rats consumed a diet enriched with the extracts the results showed an improved motor performance, and enhanced cognitive functions. In addition, the rats showed reduced oxidative stress and inflammation, and enhanced neurogenesis, Nrf2 and anti-oxidant expression. The effect of GF extracts on the augmentation of memory and learning is significant and may function through the modulation of antioxidant enzymes, signaling pathways and additional mechanisms to improve the aging process. These studies further support the recommendation of USDA for the consumption of fruits and vegetables to improve healthy aging.
Collapse
Affiliation(s)
- Jin Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Hong Zhu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | | | - Saeid Taheri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Mark S Kindy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA.,James A. Haley VA Medical Center, Tampa, FL, USA.,Shriners Hospital for Children, Tampa, FL, USA
| |
Collapse
|
33
|
|
34
|
Milenkovic I, Jarc J, Dassler E, Aronica E, Iyer A, Adle-Biassette H, Scharrer A, Reischer T, Hainfellner JA, Kovacs GG. The physiological phosphorylation of tau is critically changed in fetal brains of individuals with Down syndrome. Neuropathol Appl Neurobiol 2017; 44:314-327. [PMID: 28455903 DOI: 10.1111/nan.12406] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 03/17/2017] [Accepted: 04/15/2017] [Indexed: 01/15/2023]
Abstract
AIMS Down syndrome (DS) is a common cause of mental retardation accompanied by cognitive impairment. Comprehensive studies suggested a link between development and ageing, as nearly all individuals with DS develop Alzheimer disease (AD)-like pathology. However, there is still a paucity of data on tau in early DS to support this notion. METHODS Using morphometric immunohistochemistry we compared tau phosphorylation in normal brains and in brains of individuals with DS from early development until early postnatal life. RESULTS We observed in DS a critical loss of physiological phosphorylation of tau. Rhombencephalic structures showed prominent differences between controls and DS using antibodies AT8 (Ser-202/Thr-205) and AT180 (Thr-231). In contrast, in the subiculum only a small portion of controls deviated from DS using antibodies AT100 (Thr-212/Ser-214) and AT270 (Thr-181). With exception of the subiculum, phosphorylation-independent tau did not differ between groups, as confirmed by immunostaining for the HT-7 antibody (epitope between 159 and 163 of the human tau) as well. DISCUSSION Our observations suggest functional tau disturbance in DS brains during development, rather than axonal loss. This supports the role of tau as a further important player in the pathophysiology of cognitive impairment in DS and related AD.
Collapse
Affiliation(s)
- I Milenkovic
- Department of Neurology, Medical University of Vienna, Vienna, Austria.,Institute of Neurology, Neurodegeneration Research Group, Medical University of Vienna, Vienna, Austria
| | - J Jarc
- Institute of Neurology, Neurodegeneration Research Group, Medical University of Vienna, Vienna, Austria
| | - E Dassler
- Institute of Neurology, Neurodegeneration Research Group, Medical University of Vienna, Vienna, Austria
| | - E Aronica
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, The Netherlands.,SEIN - Stichting Epilepsie Instellingen Nederland, Heemstede, The Netherlands.,Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, The Netherlands
| | - A Iyer
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, The Netherlands
| | - H Adle-Biassette
- Inserm U1141, Paris, France.,Univ Paris Diderot, Sorbonne Paris Cité, UMRS 676, Paris, France.,Lariboisière Hospital, APHP, Paris, France
| | - A Scharrer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - T Reischer
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - J A Hainfellner
- Institute of Neurology, Neurodegeneration Research Group, Medical University of Vienna, Vienna, Austria
| | - G G Kovacs
- Institute of Neurology, Neurodegeneration Research Group, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
35
|
Gobshtis N, Tfilin M, Wolfson M, Fraifeld VE, Turgeman G. Transplantation of mesenchymal stem cells reverses behavioural deficits and impaired neurogenesis caused by prenatal exposure to valproic acid. Oncotarget 2017; 8:17443-17452. [PMID: 28407680 PMCID: PMC5392261 DOI: 10.18632/oncotarget.15245] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 01/24/2017] [Indexed: 02/06/2023] Open
Abstract
Neurodevelopmental impairment can affect lifelong brain functions such as cognitive and social behaviour, and may contribute to aging-related changes of these functions. In the present study, we hypothesized that bone marrow-derived mesenchymal stem cells (MSC) administration may repair neurodevelopmental behavioural deficits by modulating adult hippocampal neurogenesis. Indeed, postnatal intracerebral transplantation of MSC has restored cognitive and social behaviour in mice prenatally exposed to valproic acid (VPA). MSC transplantation also restored post-developmental hippocampal neurogenesis, which was impaired in VPA-exposed mice displaying delayed differentiation and maturation of newly formed neurons in the granular cell layer of the dentate gyrus. Importantly, a statistically significant correlation was found between neuronal differentiation scores and behavioural scores, suggesting a mechanistic relation between the two. We thus conclude that post-developmental MSC administration can overcome prenatal neurodevelopmental deficits and restore cognitive and social behaviours via modulation of hippocampal adult neurogenesis.
Collapse
Affiliation(s)
- Nikolai Gobshtis
- Departments of Pre-Medical Studies & Molecular Biology, Ariel University, Ariel, Israel
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva, Israe
| | - Matanel Tfilin
- Departments of Pre-Medical Studies & Molecular Biology, Ariel University, Ariel, Israel
| | - Marina Wolfson
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva, Israe
| | - Vadim E. Fraifeld
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva, Israe
| | - Gadi Turgeman
- Departments of Pre-Medical Studies & Molecular Biology, Ariel University, Ariel, Israel
| |
Collapse
|
36
|
Sher RB. The interaction of genetics and environmental toxicants in amyotrophic lateral sclerosis: results from animal models. Neural Regen Res 2017; 12:902-905. [PMID: 28761418 PMCID: PMC5514860 DOI: 10.4103/1673-5374.208564] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease that results in the progressive death of motor neurons, leading to paralysis and eventual death. There is presently no cure for ALS, and only two drugs are available, neither of which provide significant extension of life. The wide variation in onset and progression of the disease, both in sporadic and even in strongly penetrant monogenic familial forms of ALS, indicate that in addition to background genetic variation impacting the disease process, environmental exposures are likely contributors. Epidemiological evidence worldwide implicates exposures to bacterial toxins, heavy metals, pesticides, and trauma as probable environmental factors. Here, we review current advances in gene-environment interactions in ALS animal models. We report our recent discoveries in a zebrafish model of ALS in relation to exposure to the cyanobacterial toxin BMAA, and discuss several results from mouse models that show interactions with exposure to mercury and statin drugs, both leading to acceleration of the disease process. The increasing research into this combinatorial gene-environment process is just starting, but shows early promise to uncover the underlying biochemical pathways that instigate the initial motor neuron defects and lead to their rapidly progressive dysfunction.
Collapse
Affiliation(s)
- Roger B Sher
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
37
|
Abstract
Neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and frontotemporal lobar dementia are among the most pressing problems of developed societies with aging populations. Neurons carry out essential functions such as signal transmission and network integration in the central nervous system and are the main targets of neurodegenerative disease. In this Review, I address how the neuron's environment also contributes to neurodegeneration. Maintaining an optimal milieu for neuronal function rests with supportive cells termed glia and the blood-brain barrier. Accumulating evidence suggests that neurodegeneration occurs in part because the environment is affected during disease in a cascade of processes collectively termed neuroinflammation. These observations indicate that therapies targeting glial cells might provide benefit for those afflicted by neurodegenerative disorders.
Collapse
|
38
|
Abstract
Neurodevelopmental origins of functional variation in older age are increasingly being acknowledged, but identification of how early factors impact human brain and cognition throughout life has remained challenging. Much focus has been on age-specific mechanisms affecting neural foundations of cognition and their change. In contrast to this approach, we tested whether cerebral correlates of general cognitive ability (GCA) in development could be extended to the rest of the lifespan, and whether early factors traceable to prenatal stages, such as birth weight and parental education, may exert continuous influences. We measured the area of the cerebral cortex in a longitudinal sample of 974 individuals aged 4-88 y (1,633 observations). An extensive cortical region was identified wherein area related positively to GCA in development. By tracking area of the cortical region identified in the child sample throughout the lifespan, we showed that the cortical change trajectories of higher and lower GCA groups were parallel through life, suggesting continued influences of early life factors. Birth weight and parental education obtained from the Norwegian Mother-Child Cohort study were identified as such early factors of possible life-long influence. Support for a genetic component was obtained in a separate twin sample (Vietnam Era Twin Study of Aging), but birth weight in the child sample had an effect on cortical area also when controlling for possible genetic differences in terms of parental height. Our results provide novel evidence for stability in brain-cognition relationships throughout life, and indicate that early life factors impact brain and cognition for the entire life course.
Collapse
|
39
|
Gygli PE, Chang JC, Gokozan HN, Catacutan FP, Schmidt TA, Kaya B, Goksel M, Baig FS, Chen S, Griveau A, Michowski W, Wong M, Palanichamy K, Sicinski P, Nelson RJ, Czeisler C, Otero JJ. Cyclin A2 promotes DNA repair in the brain during both development and aging. Aging (Albany NY) 2016; 8:1540-70. [PMID: 27425845 PMCID: PMC4993346 DOI: 10.18632/aging.100990] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/13/2016] [Indexed: 12/24/2022]
Abstract
Various stem cell niches of the brain have differential requirements for Cyclin A2. Cyclin A2 loss results in marked cerebellar dysmorphia, whereas forebrain growth is retarded during early embryonic development yet achieves normal size at birth. To understand the differential requirements of distinct brain regions for Cyclin A2, we utilized neuroanatomical, transgenic mouse, and mathematical modeling techniques to generate testable hypotheses that provide insight into how Cyclin A2 loss results in compensatory forebrain growth during late embryonic development. Using unbiased measurements of the forebrain stem cell niche, we parameterized a mathematical model whereby logistic growth instructs progenitor cells as to the cell-types of their progeny. Our data was consistent with prior findings that progenitors proliferate along an auto-inhibitory growth curve. The growth retardation inCCNA2-null brains corresponded to cell cycle lengthening, imposing a developmental delay. We hypothesized that Cyclin A2 regulates DNA repair and that CCNA2-null progenitors thus experienced lengthened cell cycle. We demonstrate that CCNA2-null progenitors suffer abnormal DNA repair, and implicate Cyclin A2 in double-strand break repair. Cyclin A2's DNA repair functions are conserved among cell lines, neural progenitors, and hippocampal neurons. We further demonstrate that neuronal CCNA2 ablation results in learning and memory deficits in aged mice.
Collapse
Affiliation(s)
- Patrick E. Gygli
- Department of Pathology, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Joshua C. Chang
- Mathematical Biosciences Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Hamza N. Gokozan
- Department of Pathology, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Fay P. Catacutan
- Department of Pathology, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Theresa A. Schmidt
- Department of Pathology, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Behiye Kaya
- Department of Pathology, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Mustafa Goksel
- Department of Pathology, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Faisal S. Baig
- Department of Pathology, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Shannon Chen
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Amelie Griveau
- Department of Pediatrics, University of California, San Francisco School of Medicine, San Francisco, CA 94143, USA
| | - Wojciech Michowski
- Department of Genetics, Harvard Medical School and Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02115, USA
| | - Michael Wong
- Department of Pediatrics, University of California, San Francisco School of Medicine, San Francisco, CA 94143, USA
| | - Kamalakannan Palanichamy
- Department of Radiation Oncology, The Ohio State University College of Medicine. Columbus, OH 43210, USA
| | - Piotr Sicinski
- Department of Genetics, Harvard Medical School and Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02115, USA
| | - Randy J. Nelson
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Catherine Czeisler
- Department of Pathology, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - José J. Otero
- Department of Pathology, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| |
Collapse
|
40
|
Tetramethylpyrazine Promotes Migration of Neural Precursor Cells via Activating the Phosphatidylinositol 3-Kinase Pathway. Mol Neurobiol 2015; 53:6526-6539. [DOI: 10.1007/s12035-015-9551-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 11/18/2015] [Indexed: 12/31/2022]
|
41
|
Abstract
Voltage-gated sodium channels (VGSCs) are responsible for the initiation and propagation of action potentials in excitable cells. VGSCs in mammalian brain are heterotrimeric complexes of α and β subunits. Although β subunits were originally termed auxiliary, we now know that they are multifunctional signaling molecules that play roles in both excitable and nonexcitable cell types and with or without the pore-forming α subunit present. β subunits function in VGSC and potassium channel modulation, cell adhesion, and gene regulation, with particularly important roles in brain development. Mutations in the genes encoding β subunits are linked to a number of diseases, including epilepsy, sudden death syndromes like SUDEP and SIDS, and cardiac arrhythmia. Although VGSC β subunit-specific drugs have not yet been developed, this protein family is an emerging therapeutic target.
Collapse
Affiliation(s)
- Heather A O'Malley
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan 48109;
| | | |
Collapse
|
42
|
Van houcke J, De Groef L, Dekeyster E, Moons L. The zebrafish as a gerontology model in nervous system aging, disease, and repair. Ageing Res Rev 2015; 24:358-68. [PMID: 26538520 DOI: 10.1016/j.arr.2015.10.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 10/14/2015] [Accepted: 10/26/2015] [Indexed: 12/12/2022]
Abstract
Considering the increasing number of elderly in the world's population today, developing effective treatments for age-related pathologies is one of the biggest challenges in modern medical research. Age-related neurodegeneration, in particular, significantly impacts important sensory, motor, and cognitive functions, seriously constraining life quality of many patients. Although our understanding of the causal mechanisms of aging has greatly improved in recent years, animal model systems still have much to tell us about this complex process. Zebrafish (Danio rerio) have gained enormous popularity for this research topic over the past decade, since their life span is relatively short but, like humans, they are still subject to gradual aging. In addition, the extensive characterization of its well-conserved molecular and cellular physiology makes the zebrafish an excellent model to unravel the underlying mechanisms of aging, disease, and repair. This review provides a comprehensive overview of the progress made in zebrafish gerontology, with special emphasis on nervous system aging. We review the evidence that classic hallmarks of aging can also be recognized within this small vertebrate, both at the molecular and cellular level. Moreover, we illustrate the high level of similarity with age-associated human pathologies through a survey of the functional deficits that arise as zebrafish age.
Collapse
|
43
|
Sarnat HB, Philippart M, Flores-Sarnat L, Wei XC. Timing in neural maturation: arrest, delay, precociousness, and temporal determination of malformations. Pediatr Neurol 2015; 52:473-86. [PMID: 25797487 DOI: 10.1016/j.pediatrneurol.2015.01.020] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 01/29/2015] [Accepted: 01/31/2015] [Indexed: 11/30/2022]
Abstract
Timing is primordial in initiating and synchronizing each developmental process in tissue morphogenesis. Maturational arrest, delay, and precociousness all are conducive to neurological dysfunction and may determine different malformations depending on when in development the faulty timing occurred, regardless of the identification of a specific genetic mutation or an epigenetic teratogenic event. Delay and arrest are distinguished by whether further progressive development over time can be expected or the condition is static. In general, retardation of early developmental processes, such as neurulation, cellular proliferation, and migration, leads to maturational arrest. Retardation of late processes, such as synaptogenesis and myelination, are more likely to result in maturational delay. Faulty timing of neuronal maturation in relation to other developmental processes causes neurological dysfunction and abnormal electroencephalograph maturation in preterm neonates. Precocious synaptogenesis, including pruning to provide plasticity, may facilitate prenatal formation of epileptic circuitry leading to severe postnatal infantile epilepsies. The anterior commissure forms 3 weeks earlier than the corpus callosum; its presence or absence in callosal agenesis is a marker for the onset of the initial insult. An excessively thick corpus callosum may be due to delayed retraction of transitory collateral axons. Malformations that arise at different times can share a common pathogenesis with variations on the extent: timing of mitotic cycles in mosaic somatic mutations may distinguish hemimegalencephaly from focal cortical dysplasia type 2. Timing should always be considered in interpreting cerebral dysgeneses in both imaging and neuropathological diagnoses.
Collapse
Affiliation(s)
- Harvey B Sarnat
- Department of Paediatrics, University of Calgary Faculty of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada; Department of Pathology (Neuropathology), University of Calgary Faculty of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada; Department of Clinical Neurosciences, University of Calgary Faculty of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada.
| | | | - Laura Flores-Sarnat
- Department of Paediatrics, University of Calgary Faculty of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada; Department of Clinical Neurosciences, University of Calgary Faculty of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| | - Xing-Chang Wei
- Department of Paediatrics, University of Calgary Faculty of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada; Department of Radiology and Diagnostic Imaging, University of Calgary Faculty of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| |
Collapse
|
44
|
Prabowo AS, Iyer AM, Veersema TJ, Anink JJ, Schouten-van Meeteren AYN, Spliet WGM, van Rijen PC, Ferrier CH, Thom M, Aronica E. Expression of neurodegenerative disease-related proteins and caspase-3 in glioneuronal tumours. Neuropathol Appl Neurobiol 2015; 41:e1-e15. [DOI: 10.1111/nan.12143] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 04/04/2014] [Indexed: 02/06/2023]
Affiliation(s)
- A. S. Prabowo
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - A. M. Iyer
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - T. J. Veersema
- Department of Neurosurgery; University Medical Center Utrecht; Utrecht The Netherlands
- Department of Neurology; University Medical Center Utrecht; Utrecht The Netherlands
| | - J. J. Anink
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - A. Y. N. Schouten-van Meeteren
- Department of Pediatric Oncology; Emma Children's Hospital; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - W. G. M. Spliet
- Rudolf Magnus Institute for Neuroscience and Pathology; University Medical Center Utrecht; Utrecht The Netherlands
| | - P. C. van Rijen
- Department of Neurosurgery; University Medical Center Utrecht; Utrecht The Netherlands
| | - C. H. Ferrier
- Department of Neurology; University Medical Center Utrecht; Utrecht The Netherlands
- Department of Clinical Neurophysiology; University Medical Center Utrecht; Utrecht The Netherlands
| | - M. Thom
- Neuropathology Department; University College London Institute of Neurology; London UK
| | - E. Aronica
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
- Swammerdam Institute for Life Sciences; Center for Neuroscience; University of Amsterdam; Amsterdam The Netherlands
- SEIN - Stichting Epilepsie Instellingen Nederland; Heemstede The Netherlands
| |
Collapse
|
45
|
Early-Life Toxic Insults and Onset of Sporadic Neurodegenerative Diseases-an Overview of Experimental Studies. Curr Top Behav Neurosci 2015; 29:231-264. [PMID: 26695168 DOI: 10.1007/7854_2015_416] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The developmental origin of health and disease hypothesis states that adverse fetal and early childhood exposures can predispose to obesity, cardiovascular, and neurodegenerative diseases (NDDs) in adult life. Early exposure to environmental chemicals interferes with developmental programming and induces subclinical alterations that may hesitate in pathophysiology and behavioral deficits at a later life stage. The mechanisms by which perinatal insults lead to altered programming and to disease later in life are still undefined. The long latency between exposure and onset of disease, the difficulty of reconstructing early exposures, and the wealth of factors which the individual is exposed to during the life course make extremely difficult to prove the developmental origin of NDDs in clinical and epidemiological studies. An overview of animal studies assessing the long-term effects of perinatal exposure to different chemicals (heavy metals and pesticides) supports the link between exposure and hallmarks of neurodegeneration at the adult stage. Furthermore, models of maternal immune activation show that brain inflammation in early life may enhance adult vulnerability to environmental toxins, thus supporting the multiple hit hypothesis for NDDs' etiology. The study of prospective animal cohorts may help to unraveling the complex pathophysiology of sporadic NDDs. In vivo models could be a powerful tool to clarify the mechanisms through which different kinds of insults predispose to cell loss in the adult age, to establish a cause-effect relationship between "omic" signatures and disease/dysfunction later in life, and to identify peripheral biomarkers of exposure, effects, and susceptibility, for translation to prospective epidemiological studies.
Collapse
|
46
|
Watanabe K, Shibuya S, Ozawa Y, Nojiri H, Izuo N, Yokote K, Shimizu T. Superoxide dismutase 1 loss disturbs intracellular redox signaling, resulting in global age-related pathological changes. BIOMED RESEARCH INTERNATIONAL 2014; 2014:140165. [PMID: 25276767 PMCID: PMC4170698 DOI: 10.1155/2014/140165] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/29/2014] [Accepted: 08/06/2014] [Indexed: 01/14/2023]
Abstract
Aging is characterized by increased oxidative stress, chronic inflammation, and organ dysfunction, which occur in a progressive and irreversible manner. Superoxide dismutase (SOD) serves as a major antioxidant and neutralizes superoxide radicals throughout the body. In vivo studies have demonstrated that copper/zinc superoxide dismutase-deficient (Sod1(-/-)) mice show various aging-like pathologies, accompanied by augmentation of oxidative damage in organs. We found that antioxidant treatment significantly attenuated the age-related tissue changes and oxidative damage-associated p53 upregulation in Sod1(-/-) mice. This review will focus on various age-related pathologies caused by the loss of Sod1 and will discuss the molecular mechanisms underlying the pathogenesis in Sod1(-/-) mice.
Collapse
Affiliation(s)
- Kenji Watanabe
- Department of Advanced Aging Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
- Department of Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Shuichi Shibuya
- Department of Advanced Aging Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Yusuke Ozawa
- Department of Advanced Aging Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Hidetoshi Nojiri
- Department of Orthopaedics, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Naotaka Izuo
- Department of Advanced Aging Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Koutaro Yokote
- Department of Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Takahiko Shimizu
- Department of Advanced Aging Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| |
Collapse
|
47
|
Sarnat HB, Flores-Sarnat L. Morphogenesis timing of genetically programmed brain malformations in relation to epilepsy. PROGRESS IN BRAIN RESEARCH 2014; 213:181-98. [DOI: 10.1016/b978-0-444-63326-2.00010-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|