1
|
Wang Y, Ding P, Zhang K, Xu X, Li H. Correlation Between Regulation of Intestinal Flora by Danggui-Shaoyao-San and Improvement of Cognitive Impairment in Mice With Alzheimer's Disease. Brain Behav 2024; 14:e70110. [PMID: 39482855 PMCID: PMC11527834 DOI: 10.1002/brb3.70110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/21/2024] [Accepted: 10/06/2024] [Indexed: 11/03/2024] Open
Abstract
PURPOSE The abnormal central glucose metabolism in Alzheimer's disease (AD) is related to the brain-gut axis. This study aims to explore the target of Danggui-Shaoyao-San (DSS) in improving cognitive impairment. METHOD This study analyzed the differences in mice intestinal flora by 16S rRNA sequencing. The cognitive protective effects of DSS were observed through the Morris water maze and the new object recognition. The mitigation effects of DSS on Aβ and p-tau, regulatory effects on glucose metabolism targets, and intestinal structure effects were observed through brain and colon slices staining. The differences in neural ultrastructure were compared by transmission electron microscopy. FINDING The results showed that DSS affected the composition of intestinal dominant bacteria and bacteria genera and regulated the abundance of intestinal bacteria in AD mice. DSS improved the behavior of AD mice, alleviated the deposition of AD pathological products in the brain and colon, regulated the expression of glycometabolism-related proteins, and improved the colon barrier structure and neural ultrastructure in the brain of mice with AD. CONCLUSION Our findings suggest that DSS may affect AD central glucose metabolism and improve cognition by regulating the gut-brain axis.
Collapse
Affiliation(s)
- Ya‐Han Wang
- Department of NeurologyAffiliated Hospital of Shandong University of Traditional Chinese MedicineJinanChina
| | - Peng‐Li Ding
- The First Clinical Medical CollegeShandong University of Traditional Chinese MedicineJinanChina
| | - Kai‐Xin Zhang
- The First Clinical Medical CollegeShandong University of Traditional Chinese MedicineJinanChina
| | - Xiang‐Qing Xu
- Department of NeurologyAffiliated Hospital of Shandong University of Traditional Chinese MedicineJinanChina
| | - He Li
- The First Clinical Medical CollegeShandong University of Traditional Chinese MedicineJinanChina
| |
Collapse
|
2
|
Gilbreath D, Hagood D, Larson-Prior L. A Systematic Review over the Effect of Early Infant Diet on Neurodevelopment: Insights from Neuroimaging. Nutrients 2024; 16:1703. [PMID: 38892636 PMCID: PMC11174660 DOI: 10.3390/nu16111703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/29/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
The optimization of infant neuronal development through nutrition is an increasingly studied area. While human milk consumption during infancy is thought to give a slight cognitive advantage throughout early childhood in comparison to commercial formula, the biological underpinnings of this process are less well-known and debated in the literature. This systematic review seeks to quantitatively analyze whether early diet affects infant neurodevelopment as measured by various neuroimaging modalities and techniques. Results presented suggest that human milk does have a slight positive impact on the structural development of the infant brain-and that this impact is larger in preterm infants. Other diets with distinct macronutrient compositions were also considered, although these had more conflicting results.
Collapse
Affiliation(s)
- Dylan Gilbreath
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Science, Little Rock, AR 72207, USA;
- Arkansas Children’s Nutrition Center, Little Rock, AR 72202, USA;
| | - Darcy Hagood
- Arkansas Children’s Nutrition Center, Little Rock, AR 72202, USA;
| | - Linda Larson-Prior
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Science, Little Rock, AR 72207, USA;
- Arkansas Children’s Nutrition Center, Little Rock, AR 72202, USA;
| |
Collapse
|
3
|
Mercier O, Quilichini PP, Magalon K, Gil F, Ghestem A, Richard F, Boudier T, Cayre M, Durbec P. Transient demyelination causes long-term cognitive impairment, myelin alteration and network synchrony defects. Glia 2024; 72:960-981. [PMID: 38363046 DOI: 10.1002/glia.24513] [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: 05/17/2023] [Revised: 01/26/2024] [Accepted: 02/05/2024] [Indexed: 02/17/2024]
Abstract
In the adult brain, activity-dependent myelin plasticity is required for proper learning and memory consolidation. Myelin loss, alteration, or even subtle structural modifications can therefore compromise the network activity, leading to functional impairment. In multiple sclerosis, spontaneous myelin repair process is possible, but it is heterogeneous among patients, sometimes leading to functional recovery, often more visible at the motor level than at the cognitive level. In cuprizone-treated mouse model, massive brain demyelination is followed by spontaneous and robust remyelination. However, reformed myelin, although functional, may not exhibit the same morphological characteristics as developmental myelin, which can have an impact on the activity of neural networks. In this context, we used the cuprizone-treated mouse model to analyze the structural, functional, and cognitive long-term effects of transient demyelination. Our results show that an episode of demyelination induces despite remyelination long-term cognitive impairment, such as deficits in spatial working memory, social memory, cognitive flexibility, and hyperactivity. These deficits were associated with a reduction in myelin content in the medial prefrontal cortex (mPFC) and hippocampus (HPC), as well as structural myelin modifications, suggesting that the remyelination process may be imperfect in these structures. In vivo electrophysiological recordings showed that the demyelination episode altered the synchronization of HPC-mPFC activity, which is crucial for memory processes. Altogether, our data indicate that the myelin repair process following transient demyelination does not allow the complete recovery of the initial myelin properties in cortical structures. These subtle modifications alter network features, leading to prolonged cognitive deficits in mice.
Collapse
Affiliation(s)
- Océane Mercier
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Pascale P Quilichini
- U1106 after INS, Aix Marseille Univ, INSERM, INS, Inst Neurosci Syst, Marseille, France
| | - Karine Magalon
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Florian Gil
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Antoine Ghestem
- U1106 after INS, Aix Marseille Univ, INSERM, INS, Inst Neurosci Syst, Marseille, France
| | - Fabrice Richard
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Thomas Boudier
- Aix Marseille Univ, Turing Centre for Living Systems, Marseille, France
| | - Myriam Cayre
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Pascale Durbec
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| |
Collapse
|
4
|
Poggi G, Klaus F, Pryce CR. Pathophysiology in cortico-amygdala circuits and excessive aversion processing: the role of oligodendrocytes and myelination. Brain Commun 2024; 6:fcae140. [PMID: 38712320 PMCID: PMC11073757 DOI: 10.1093/braincomms/fcae140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/27/2023] [Accepted: 04/16/2024] [Indexed: 05/08/2024] Open
Abstract
Stress-related psychiatric illnesses, such as major depressive disorder, anxiety and post-traumatic stress disorder, present with alterations in emotional processing, including excessive processing of negative/aversive stimuli and events. The bidirectional human/primate brain circuit comprising anterior cingulate cortex and amygdala is of fundamental importance in processing emotional stimuli, and in rodents the medial prefrontal cortex-amygdala circuit is to some extent analogous in structure and function. Here, we assess the comparative evidence for: (i) Anterior cingulate/medial prefrontal cortex<->amygdala bidirectional neural circuits as major contributors to aversive stimulus processing; (ii) Structural and functional changes in anterior cingulate cortex<->amygdala circuit associated with excessive aversion processing in stress-related neuropsychiatric disorders, and in medial prefrontal cortex<->amygdala circuit in rodent models of chronic stress-induced increased aversion reactivity; and (iii) Altered status of oligodendrocytes and their oligodendrocyte lineage cells and myelination in anterior cingulate/medial prefrontal cortex<->amygdala circuits in stress-related neuropsychiatric disorders and stress models. The comparative evidence from humans and rodents is that their respective anterior cingulate/medial prefrontal cortex<->amygdala circuits are integral to adaptive aversion processing. However, at the sub-regional level, the anterior cingulate/medial prefrontal cortex structure-function analogy is incomplete, and differences as well as similarities need to be taken into account. Structure-function imaging studies demonstrate that these neural circuits are altered in both human stress-related neuropsychiatric disorders and rodent models of stress-induced increased aversion processing. In both cases, the changes include altered white matter integrity, albeit the current evidence indicates that this is decreased in humans and increased in rodent models. At the cellular-molecular level, in both humans and rodents, the current evidence is that stress disorders do present with changes in oligodendrocyte lineage, oligodendrocytes and/or myelin in these neural circuits, but these changes are often discordant between and even within species. Nonetheless, by integrating the current comparative evidence, this review provides a timely insight into this field and should function to inform future studies-human, monkey and rodent-to ascertain whether or not the oligodendrocyte lineage and myelination are causally involved in the pathophysiology of stress-related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Giulia Poggi
- Preclinical Laboratory for Translational Research into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, CH-8008 Zurich, Switzerland
| | - Federica Klaus
- Department of Psychiatry, University of California San Diego, San Diego, CA 92093, USA
- Desert-Pacific Mental Illness Research Education and Clinical Center, VA San Diego Healthcare System, San Diego, CA 92093, USA
| | - Christopher R Pryce
- Preclinical Laboratory for Translational Research into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, CH-8008 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, 8057 Zurich, Switzerland
- URPP Adaptive Brain Circuits in Development and Learning (AdaBD), University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
5
|
López-Murillo C, Hinestroza-Morales S, Henny P, Toledo J, Cardona-Gómez GP, Rivera-Gutiérrez H, Posada-Duque R. Differences in vocal brain areas and astrocytes between the house wren and the rufous-tailed hummingbird. Front Neuroanat 2024; 18:1339308. [PMID: 38601797 PMCID: PMC11004282 DOI: 10.3389/fnana.2024.1339308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 03/06/2024] [Indexed: 04/12/2024] Open
Abstract
The house wren shows complex song, and the rufous-tailed hummingbird has a simple song. The location of vocal brain areas supports the song's complexity; however, these still need to be studied. The astrocytic population in songbirds appears to be associated with change in vocal control nuclei; however, astrocytic distribution and morphology have not been described in these species. Consequently, we compared the distribution and volume of the vocal brain areas: HVC, RA, Area X, and LMAN, cell density, and the morphology of astrocytes in the house wren and the rufous-tailed hummingbird. Individuals of the two species were collected, and their brains were analyzed using serial Nissl- NeuN- and MAP2-stained tissue scanner imaging, followed by 3D reconstructions of the vocal areas; and GFAP and S100β astrocytes were analyzed in both species. We found that vocal areas were located close to the cerebral midline in the house wren and a more lateralized position in the rufous-tailed hummingbird. The LMAN occupied a larger volume in the rufous-tailed hummingbird, while the RA and HVC were larger in the house wren. While Area X showed higher cell density in the house wren than the rufous-tailed hummingbird, the LMAN showed a higher density in the rufous-tailed hummingbird. In the house wren, GFAP astrocytes in the same bregma where the vocal areas were located were observed at the laminar edge of the pallium (LEP) and in the vascular region, as well as in vocal motor relay regions in the pallidum and mesencephalon. In contrast, GFAP astrocytes were found in LEP, but not in the pallidum and mesencephalon in hummingbirds. Finally, when comparing GFAP astrocytes in the LEP region of both species, house wren astrocytes exhibited significantly more complex morphology than those of the rufous-tailed hummingbird. These findings suggest a difference in the location and cellular density of vocal circuits, as well as morphology of GFAP astrocytes between the house wren and the rufous-tailed hummingbird.
Collapse
Affiliation(s)
- Carolina López-Murillo
- Área de Neurofisiología Celular, Grupo de Neurociencias de Antioquia, Instituto de Biología, Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia, Medellin, Colombia
| | - Santiago Hinestroza-Morales
- Área de Neurofisiología Celular, Grupo de Neurociencias de Antioquia, Instituto de Biología, Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia, Medellin, Colombia
| | - Pablo Henny
- Laboratorio de Neuroanatomía, Departamento de Anatomía, and Centro Interdisciplinario de Neurociencia, NeuroUC, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge Toledo
- Scientific Equipment Network REDECA, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Gloria Patricia Cardona-Gómez
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de Antioquia, Facultad de Medicina, Sede de Investigaciones Universitarias, Universidad de Antioquia, Medellin, Colombia
| | - Héctor Rivera-Gutiérrez
- Grupo de Investigación de Ecología y Evolución de Vertebrados, Instituto de Biología, Universidad de Antioquia, Medellin, Colombia
| | - Rafael Posada-Duque
- Área de Neurofisiología Celular, Grupo de Neurociencias de Antioquia, Instituto de Biología, Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia, Medellin, Colombia
| |
Collapse
|
6
|
Nazarova A, Drobinin V, Helmick CA, Schmidt MH, Cookey J, Uher R. Intracortical Myelin in Youths at Risk for Depression. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100285. [PMID: 38323155 PMCID: PMC10844807 DOI: 10.1016/j.bpsgos.2023.100285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/28/2023] [Accepted: 12/02/2023] [Indexed: 02/08/2024] Open
Abstract
Background Major depressive disorder (MDD) is a leading cause of disability. To understand why depression develops, it is important to distinguish between early neural markers of vulnerability that precede the onset of MDD and features that develop during depression. Recent neuroimaging findings suggest that reduced global and regional intracortical myelination (ICM), especially in the lateral prefrontal cortex, may be associated with depression, but it is unknown whether it is a precursor or a consequence of MDD. The study of offspring of affected parents offers the opportunity to distinguish between precursors and consequences by examining individuals who carry high risk at a time when they have not experienced depression. Methods We acquired 129 T1-weighted and T2-weighted scans from 56 (25 female) unaffected offspring of parents with depression and 114 scans from 63 (34 female) unaffected offspring of parents without a history of depression (ages 9 to 16 years). To assess scan quality, we calculated test-retest reliability. We used the scan ratios to calculate myelin maps for 68 cortical regions. We analyzed data using mixed-effects modeling. Results ICM did not differ between high and low familial risk youths in global (B = 0.06, SE = 0.03, p = .06) or regional (B = 0.05, SE = 0.03, p = .08) analyses. Our pediatric sample had high ICM reliability (intraclass correlation coefficient = 0.79; 95% CI, 0.55-0.88). Conclusions Based on our results, reduced ICM does not appear to be a precursor of MDD. Future studies should examine ICM in familial high-risk youths across a broad developmental period.
Collapse
Affiliation(s)
- Anna Nazarova
- Department of Psychiatry, Dalhousie University, Abbie J. Lane Memorial Building Queen Elizabeth II Health Sciences Centre, Halifax, Nova Scotia, Canada
- Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| | - Vladislav Drobinin
- Department of Psychiatry, Dalhousie University, Abbie J. Lane Memorial Building Queen Elizabeth II Health Sciences Centre, Halifax, Nova Scotia, Canada
| | - Carl A. Helmick
- Department of Psychiatry, Dalhousie University, Abbie J. Lane Memorial Building Queen Elizabeth II Health Sciences Centre, Halifax, Nova Scotia, Canada
| | - Matthias H. Schmidt
- Department of Diagnostic Radiology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jacob Cookey
- Department of Psychiatry, Dalhousie University, Abbie J. Lane Memorial Building Queen Elizabeth II Health Sciences Centre, Halifax, Nova Scotia, Canada
- Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| | - Rudolf Uher
- Department of Psychiatry, Dalhousie University, Abbie J. Lane Memorial Building Queen Elizabeth II Health Sciences Centre, Halifax, Nova Scotia, Canada
- Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| |
Collapse
|
7
|
Kambe J, Usuda K, Inoue R, Hirayama K, Ito M, Suenaga K, Masukado S, Liu H, Miyata S, Li C, Kimura I, Yamamoto Y, Nagaoka K. Hydrogen peroxide in breast milk is crucial for gut microbiota formation and myelin development in neonatal mice. Gut Microbes 2024; 16:2359729. [PMID: 38816999 PMCID: PMC11146441 DOI: 10.1080/19490976.2024.2359729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 05/21/2024] [Indexed: 06/01/2024] Open
Abstract
Early life environment influences mammalian brain development, a growing area of research within the Developmental Origins of Health and Disease framework, necessitating a deeper understanding of early life factors on children's brain development. This study introduces a mouse model, LAO1 knockout mice, to investigate the relationship between breast milk, the gut microbiome, and brain development. The results reveal that breast milk's reactive oxygen species (ROS) are vital in shaping the neonatal gut microbiota. Decreased hydrogen peroxide (H2O2) levels in milk disrupt the gut microbiome and lead to abnormal metabolite production, including D-glucaric acid. This metabolite inhibits hippocampal myelin formation during infancy, potentially contributing to behavioral abnormalities observed in adulthood. These findings suggest that H2O2 in breast milk is crucial for normal gut microbiota formation and brain development, with implications for understanding and potentially treating neurodevelopmental disorders in humans.
Collapse
Affiliation(s)
- Jun Kambe
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Kento Usuda
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Ryo Inoue
- Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University, Osaka, Japan
| | - Kazuhiko Hirayama
- Laboratory of Veterinary Public Health, Department of Veterinary Medical Science, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Masahiko Ito
- Department of Virology and Parasitology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Ken Suenaga
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Sora Masukado
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Hong Liu
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Shiho Miyata
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Chunmei Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Ikuo Kimura
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Yuki Yamamoto
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Kentaro Nagaoka
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| |
Collapse
|
8
|
Zhuang H, Li Q, Sun C, Xu D, Gan G, Zhang C, Chen C, Yuan Y, Liu L, Xiao Y, Yao X, Wang C, Kang X, Yang C, Zhao J, Chen W, Wang J, Li J, Luo C, Wang J, Jia X, Yu Z, Liu L. Voluntary wheel exercise ameliorates cognitive impairment, hippocampal neurodegeneration and microglial abnormalities preceded by demyelination in a male mouse model of noise-induced hearing loss. Brain Behav Immun 2023; 114:325-348. [PMID: 37683962 DOI: 10.1016/j.bbi.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/23/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023] Open
Abstract
Acquired peripheral hearing loss (APHL) in midlife has been identified as the greatest modifiable risk factor for dementia; however, the pathophysiological neural mechanisms linking APHL with an increased risk of dementia remain to be elucidated. Here, in an adult male mouse model of noise-induced hearing loss (NIHL), one of the most common forms of APHL, we demonstrated accelerated age-related cognitive decline and hippocampal neurodegeneration during a 6-month follow-up period, accompanied by progressive hippocampal microglial aberrations preceded by immediate-onset transient elevation in serum glucocorticoids and delayed-onset sustained myelin disruption in the hippocampus. Pretreatment with the glucocorticoid receptor antagonist RU486 before stressful noise exposure partially mitigated the early activation of hippocampal microglia, which were present at 7 days post noise exposure (7DPN), but had no impact on later microglial aberrations, hippocampal neurodegeneration, or cognitive decline exhibited at 1 month post noise exposure (1MPN). One month of voluntary wheel exercise following noise exposure barely affected either the hearing threshold shift or hippocampal myelin changes but effectively countered cognitive impairment and the decline in hippocampal neurogenesis in NIHL mice at 1MPN, paralleled by the normalization of microglial morphology, which coincided with a reduction in microglial myelin inclusions and a restoration of microglial hypoxia-inducible factor-1α (HIF1α) expression. Our results indicated that accelerated cognitive deterioration and hippocampal neuroplastic decline following NIHL are most likely driven by the maladaptive response of hippocampal microglia to myelin damage secondary to hearing loss, and we also demonstrated the potential of voluntary physical exercise as a promising and cost-effective strategy to alleviate the detrimental impact of APHL on cognitive function and thus curtail the high and continuously increasing global burden of dementia. Furthermore, the findings of the present study highlight the contribution of myelin debris overload to microglial malfunction and identify the microglial HIF1α-related pathway as an attractive candidate for future comprehensive investigation to obtain a more definitive picture of the underlying mechanisms linking APHL and dementia.
Collapse
Affiliation(s)
- Hong Zhuang
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Qian Li
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Congli Sun
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Dan Xu
- School of Public Health, Southeast University, Nanjing 210009, China
| | - Guangming Gan
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Chenchen Zhang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Chen Chen
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Yang Yuan
- Department of Endocrinology, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China
| | - Linchen Liu
- Department of Rheumatology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yu Xiao
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiuting Yao
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Conghui Wang
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiaoming Kang
- School of Life Science and Technology, Southeast University, Nanjing 210009, China
| | - Chenxi Yang
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jingyi Zhao
- School of Life Science and Technology, Southeast University, Nanjing 210009, China
| | - Wenhao Chen
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Jiatang Wang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Jinyu Li
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Caichen Luo
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Jie Wang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Xirui Jia
- School of Life Science and Technology, Southeast University, Nanjing 210009, China
| | - Zhehao Yu
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Lijie Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
9
|
Dong X, Hong H, Cui Z. Function of GSK‑3 signaling in spinal cord injury (Review). Exp Ther Med 2023; 26:541. [PMID: 37869638 PMCID: PMC10587879 DOI: 10.3892/etm.2023.12240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 08/10/2023] [Indexed: 10/24/2023] Open
Abstract
Spinal cord injury (SCI) is a major social problem with a heavy burden on patient physiology and psychology. Glial scar formation and irreversible neuron loss are the two key points during SCI progression. During the acute phase of spinal cord injury, glial scars form, limiting the progression of inflammation. However, in the subacute or chronic phase, glial scarring inhibits axon regeneration. Following spinal cord injury, irreversible loss of neurons leads to further aggravation of spinal cord injury. Several therapies have been developed to improve either glial scar or neuron loss; however, few therapies reach the stage of clinical trials and there are no mainstream therapies for SCI. Exploring the key mechanism of SCI is crucial for finding further treatments. Glycogen synthase kinase-3 (GSK-3) is a widely expressed kinase with important physiological and pathophysiological functions in vivo. Dysfunction of the GSK-3 signaling pathway during SCI has been widely discussed for controlling neurite growth in vitro and in vivo, improving the proliferation and neuronal differentiation of endogenous neural stem cells and functional recovery from spinal cord injury. SCI can decrease the phosphorylated (p)/total (t)-GSK-3β ratio, which leads to an increase in apoptosis, whereas treatment with GSK-3 inhibitors can promote neurogenesis. In addition, several therapies for the treatment of SCI involve signaling pathways associated with GSK-3. Furthermore, signaling pathways associated with GSK-3 also participate in the pathological process of neuropathic pain that remains following SCI. The present review summarized the roles of GSK-3 signaling in SCI to aid in the understanding of GSK-3 signaling during the pathological processes of SCI and to provide evidence for the development of comprehensive treatments.
Collapse
Affiliation(s)
- Xiong Dong
- Department of Spinal Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Hongxiang Hong
- Department of Spinal Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Zhiming Cui
- Department of Spinal Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
10
|
Chen K, Cambi F, Kozai TDY. Pro-myelinating clemastine administration improves recording performance of chronically implanted microelectrodes and nearby neuronal health. Biomaterials 2023; 301:122210. [PMID: 37413842 PMCID: PMC10528716 DOI: 10.1016/j.biomaterials.2023.122210] [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: 02/06/2023] [Revised: 06/08/2023] [Accepted: 06/19/2023] [Indexed: 07/08/2023]
Abstract
Intracortical microelectrodes have become a useful tool in neuroprosthetic applications in the clinic and to understand neurological disorders in basic neurosciences. Many of these brain-machine interface technology applications require successful long-term implantation with high stability and sensitivity. However, the intrinsic tissue reaction caused by implantation remains a major failure mechanism causing loss of recorded signal quality over time. Oligodendrocytes remain an underappreciated intervention target to improve chronic recording performance. These cells can accelerate action potential propagation and provides direct metabolic support for neuronal health and functionality. However, implantation injury causes oligodendrocyte degeneration and leads to progressive demyelination in surrounding brain tissue. Previous work highlighted that healthy oligodendrocytes are necessary for greater electrophysiological recording performance and the prevention of neuronal silencing around implanted microelectrodes over the chronic implantation period. Thus, we hypothesize that enhancing oligodendrocyte activity with a pharmaceutical drug, Clemastine, will prevent the chronic decline of microelectrode recording performance. Electrophysiological evaluation showed that the promyelination Clemastine treatment significantly elevated the signal detectability and quality, rescued the loss of multi-unit activity, and increased functional interlaminar connectivity over 16-weeks of implantation. Additionally, post-mortem immunohistochemistry showed that increased oligodendrocyte density and myelination coincided with increased survival of both excitatory and inhibitory neurons near the implant. Overall, we showed a positive relationship between enhanced oligodendrocyte activity and neuronal health and functionality near the chronically implanted microelectrode. This study shows that therapeutic strategy that enhance oligodendrocyte activity is effective for integrating the functional device interface with brain tissue over chronic implantation period.
Collapse
Affiliation(s)
- Keying Chen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Franca Cambi
- Veterans Administration Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA.
| |
Collapse
|
11
|
Yu M, Wang Z, Wang D, Aierxi M, Ma Z, Wang Y. Oxidative stress following spinal cord injury: From molecular mechanisms to therapeutic targets. J Neurosci Res 2023; 101:1538-1554. [PMID: 37272728 DOI: 10.1002/jnr.25221] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 05/12/2023] [Accepted: 05/18/2023] [Indexed: 06/06/2023]
Abstract
Spinal cord injury (SCI) is a medical condition that results from severe trauma to the central nervous system; it imposes great psychological and economic burdens on affected patients and their families. The dynamic balance between reactive oxygen species (ROS) and antioxidants is essential for maintaining normal cellular physiological functions. As important intracellular signaling molecules, ROS regulate numerous physiological activities, including vascular reactivity and neuronal function. However, excessive ROS can cause damage to cellular macromolecules, including DNA, lipids, and proteins; this damage eventually leads to cell death. This review discusses the mechanisms of oxidative stress in SCI and describes some signaling pathways that regulate oxidative injury after injury, with the aim of providing guidance for the development of novel SCI treatment strategies.
Collapse
Affiliation(s)
- Mengsi Yu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Zhiying Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Dongmin Wang
- Medical College of Northwest Minzu University, Lanzhou, China
| | - Milikemu Aierxi
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Zhanjun Ma
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, UCLouvain, Brussels, Belgium
| | - Yonggang Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
12
|
Mishra E, Thakur MK. Mdivi-1 Rescues Memory Decline in Scopolamine-Induced Amnesic Male Mice by Ameliorating Mitochondrial Dynamics and Hippocampal Plasticity. Mol Neurobiol 2023; 60:5426-5449. [PMID: 37314656 DOI: 10.1007/s12035-023-03397-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/22/2023] [Indexed: 06/15/2023]
Abstract
Memory loss, often known as amnesia, is common in the elderly population and refers to forgetting facts and experiences. It is associated with increased mitochondrial fragmentation, though the contribution of mitochondrial dynamics in amnesia is poorly understood. Therefore, the present study is aimed at elucidating the role of Mdivi-1 in mitochondrial dynamics, hippocampal plasticity, and memory during scopolamine (SC)-induced amnesia. The findings imply that Mdivi-1 significantly increased the expression of Arc and BDNF proteins in the hippocampus of SC-induced amnesic mice, validating improved recognition and spatial memory. Moreover, an improved mitochondrial ultrastructure was attributed to a decline in the percentage of fragmented and spherical-shaped mitochondria after Mdivi-1 treatment in SC-induced mice. The significant downregulation of p-Drp1 (S616) protein and upregulation of Mfn2, LC3BI, and LC3BII proteins in Mdivi-1-treated SC-induced mice indicated a decline in fragmented mitochondrial number and healthy mitochondrial dynamics. Mdivi-1 treatment alleviated ROS production and Caspase-3 activity and elevated mitochondrial membrane potential, Vdac1 expression, ATP production, and myelination, resulting in reduced neurodegeneration in SC mice. Furthermore, the decline of pro-apoptotic protein cytochrome-c and increase of anti-apoptotic proteins Procaspase-9 and Bcl-2 in Mdivi-1-treated SC-induced mice suggested improved neuronal health. Mdivi-1 also increased the dendritic arborization and spine density, which was further corroborated by increased expression of synaptophysin and PSD95. In conclusion, the current study suggests that Mdivi-1 treatment improves mitochondrial ultrastructure and function through the regulation of mitochondrial dynamics. These changes further improve neuronal cell density, myelination, dendritic arborization, and spine density, decrease neurodegeneration, and improve recognition and spatial memory. Schematic presentation depicts that Mdivi-1 rescues memory decline in scopolamine-induced amnesic male mice by ameliorating mitochondrial dynamics and hippocampal plasticity.
Collapse
Affiliation(s)
- Ela Mishra
- Biochemistry and Molecular Biology Laboratory, Centre of Advanced Study, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221 005, India
| | - Mahendra Kumar Thakur
- Biochemistry and Molecular Biology Laboratory, Centre of Advanced Study, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221 005, India.
| |
Collapse
|
13
|
Soman SM, Vijayakumar N, Thomson P, Ball G, Hyde C, Silk TJ. Cortical structural and functional coupling during development and implications for attention deficit hyperactivity disorder. Transl Psychiatry 2023; 13:252. [PMID: 37433763 DOI: 10.1038/s41398-023-02546-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/13/2023] Open
Abstract
Functional connectivity is scaffolded by the structural connections of the brain. Disruptions of either structural or functional connectivity can lead to deficits in cognitive functions and increase the risk for neurodevelopmental disorders such as attention deficit hyperactivity disorder (ADHD). To date, very little research has examined the association between structural and functional connectivity in typical development, while no studies have attempted to understand the development of structure-function coupling in children with ADHD. 175 individuals (84 typically developing children and 91 children with ADHD) participated in a longitudinal neuroimaging study with up to three waves. In total, we collected 278 observations between the ages 9 and 14 (139 each in typically developing controls and ADHD). Regional measures of structure-function coupling were calculated at each timepoint using Spearman's rank correlation and mixed effect models were used to determine group differences and longitudinal changes in coupling over time. In typically developing children, we observed increases in structure-function coupling strength across multiple higher-order cognitive and sensory regions. Overall, weaker coupling was observed in children with ADHD, mainly in the prefrontal cortex, superior temporal gyrus, and inferior parietal cortex. Further, children with ADHD showed an increased rate of coupling strength predominantly in the inferior frontal gyrus, superior parietal cortex, precuneus, mid-cingulate, and visual cortex, compared to no corresponding change over time in typically developing controls. This study provides evidence of the joint maturation of structural and functional brain connections in typical development across late childhood to mid-adolescence, particularly in regions that support cognitive maturation. Findings also suggest that children with ADHD exhibit different patterns of structure-function coupling, suggesting atypical patterns of coordinated white matter and functional connectivity development predominantly in the regions overlapping with the default mode network, salience network, and dorsal attention network during late childhood to mid-adolescence.
Collapse
Affiliation(s)
- Shania Mereen Soman
- Centre for Social and Early Emotional Development and School of Psychology, Deakin University, Burwood, VIC, 3125, Australia.
| | - Nandita Vijayakumar
- Centre for Social and Early Emotional Development and School of Psychology, Deakin University, Burwood, VIC, 3125, Australia
| | - Phoebe Thomson
- Child Mind Institute, New York, NY, 10022, USA
- Department of Paediatrics, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Gareth Ball
- Department of Paediatrics, University of Melbourne, Parkville, VIC, 3010, Australia
- Developmental Imaging, Murdoch Children's Research Institute, Flemington Road, Parkville, VIC, 3052, Australia
| | - Christian Hyde
- Centre for Social and Early Emotional Development and School of Psychology, Deakin University, Burwood, VIC, 3125, Australia
| | - Timothy J Silk
- Centre for Social and Early Emotional Development and School of Psychology, Deakin University, Burwood, VIC, 3125, Australia.
- Developmental Imaging, Murdoch Children's Research Institute, Flemington Road, Parkville, VIC, 3052, Australia.
| |
Collapse
|
14
|
Wang W, Bo T, Zhang G, Li J, Ma J, Ma L, Hu G, Tong H, Lv Q, Araujo DJ, Luo D, Chen Y, Wang M, Wang Z, Wang GZ. Noncoding transcripts are linked to brain resting-state activity in non-human primates. Cell Rep 2023; 42:112652. [PMID: 37335775 DOI: 10.1016/j.celrep.2023.112652] [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: 09/21/2022] [Revised: 04/05/2023] [Accepted: 05/30/2023] [Indexed: 06/21/2023] Open
Abstract
Brain-derived transcriptomes are known to correlate with resting-state brain activity in humans. Whether this association holds in nonhuman primates remains uncertain. Here, we search for such molecular correlates by integrating 757 transcriptomes derived from 100 macaque cortical regions with resting-state activity in separate conspecifics. We observe that 150 noncoding genes explain variations in resting-state activity at a comparable level with protein-coding genes. In-depth analysis of these noncoding genes reveals that they are connected to the function of nonneuronal cells such as oligodendrocytes. Co-expression network analysis finds that the modules of noncoding genes are linked to both autism and schizophrenia risk genes. Moreover, genes associated with resting-state noncoding genes are highly enriched in human resting-state functional genes and memory-effect genes, and their links with resting-state functional magnetic resonance imaging (fMRI) signals are altered in the brains of patients with autism. Our results highlight the potential for noncoding RNAs to explain resting-state activity in the nonhuman primate brain.
Collapse
Affiliation(s)
- Wei Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Tingting Bo
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Clinical Neuroscience Center, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ge Zhang
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, Henan, China
| | - Jie Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Junjie Ma
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Liangxiao Ma
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ganlu Hu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Huige Tong
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qian Lv
- School of Psychological and Cognitive Sciences, Beijing Key Laboratory of Behavior and Mental Health, IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Daniel J Araujo
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Dong Luo
- School of Biomedical Engineering, Hainan University, Haikou, Hainan, China
| | - Yuejun Chen
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China
| | - Meiyun Wang
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, Henan, China
| | - Zheng Wang
- School of Psychological and Cognitive Sciences, Beijing Key Laboratory of Behavior and Mental Health, IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China; School of Biomedical Engineering, Hainan University, Haikou, Hainan, China.
| | - Guang-Zhong Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
15
|
Ścibior A, Llopis J, Dobrakowski PP, Męcik-Kronenberg T. CNS-Related Effects Caused by Vanadium at Realistic Exposure Levels in Humans: A Comprehensive Overview Supplemented with Selected Animal Studies. Int J Mol Sci 2023; 24:ijms24109004. [PMID: 37240351 DOI: 10.3390/ijms24109004] [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: 04/27/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Neurodegenerative disorders, which are currently incurable diseases of the nervous system, are a constantly growing social concern. They are progressive and lead to gradual degeneration and/or death of nerve cells, resulting in cognitive deterioration or impaired motor functions. New therapies that would ensure better treatment results and contribute to a significant slowdown in the progression of neurodegenerative syndromes are constantly being sought. Vanadium (V), which is an element with a wide range of impacts on the mammalian organism, is at the forefront among the different metals studied for their potential therapeutic use. On the other hand, it is a well-known environmental and occupational pollutant and can exert adverse effects on human health. As a strong pro-oxidant, it can generate oxidative stress involved in neurodegeneration. Although the detrimental effects of vanadium on the CNS are relatively well recognized, the role of this metal in the pathophysiology of various neurological disorders, at realistic exposure levels in humans, is not yet well characterized. Hence, the main goal of this review is to summarize data on the neurological side effects/neurobehavioral alterations in humans, in relation to vanadium exposure, with the focus on the levels of this metal in biological fluids/brain tissues of subjects with some neurodegenerative syndromes. Data collected in the present review indicate that vanadium cannot be excluded as a factor playing a pivotal role in the etiopathogenesis of neurodegenerative illnesses, and point to the need for additional extensive epidemiological studies that will provide more evidence supporting the relationship between vanadium exposure and neurodegeneration in humans. Simultaneously, the reviewed data, clearly showing the environmental impact of vanadium on health, suggest that more attention should be paid to chronic diseases related to vanadium and to the assessment of the dose-response relationship.
Collapse
Affiliation(s)
- Agnieszka Ścibior
- Laboratory of Oxidative Stress, Department of Biomedicine and Environmental Research, Institute of Biological Sciences, Faculty of Medicine, The John Paul II Catholic University of Lublin, Konstantynów St. 1J, 20-708 Lublin, Poland
| | - Juan Llopis
- Department of Physiology, Institute of Nutrition and Food Technology ''José Mataix", Biomedical Research Centre, University of Granada, 18100 Armilla, Spain
- Sport and Health Research Centre, University of Granada, 18016 Granada, Spain
| | - Paweł Piotr Dobrakowski
- Psychology Institute, Humanitas University in Sosnowiec, Jana Kilińskiego St. 43, 41-200 Sosnowiec, Poland
| | - Tomasz Męcik-Kronenberg
- Department of Pathomorphology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 3 Maja St. 13, 41-800 Zabrze, Poland
| |
Collapse
|
16
|
Maitre M, Jeltsch-David H, Okechukwu NG, Klein C, Patte-Mensah C, Mensah-Nyagan AG. Myelin in Alzheimer's disease: culprit or bystander? Acta Neuropathol Commun 2023; 11:56. [PMID: 37004127 PMCID: PMC10067200 DOI: 10.1186/s40478-023-01554-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with neuronal and synaptic losses due to the accumulation of toxic amyloid β (Αβ) peptide oligomers, plaques, and tangles containing tau (tubulin-associated unit) protein. While familial AD is caused by specific mutations, the sporadic disease is more common and appears to result from a complex chronic brain neuroinflammation with mitochondriopathies, inducing free radicals' accumulation. In aged brain, mutations in DNA and several unfolded proteins participate in a chronic amyloidosis response with a toxic effect on myelin sheath and axons, leading to cognitive deficits and dementia. Αβ peptides are the most frequent form of toxic amyloid oligomers. Accumulations of misfolded proteins during several years alters different metabolic mechanisms, induce chronic inflammatory and immune responses with toxic consequences on neuronal cells. Myelin composition and architecture may appear to be an early target for the toxic activity of Aβ peptides and others hydrophobic misfolded proteins. In this work, we describe the possible role of early myelin alterations in the genesis of neuronal alterations and the onset of symptomatology. We propose that some pathophysiological and clinical forms of the disease may arise from structural and metabolic disorders in the processes of myelination/demyelination of brain regions where the accumulation of non-functional toxic proteins is important. In these forms, the primacy of the deleterious role of amyloid peptides would be a matter of questioning and the initiating role of neuropathology would be primarily the fact of dysmyelination.
Collapse
Affiliation(s)
- Michel Maitre
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France.
| | - Hélène Jeltsch-David
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
- Biotechnologie et signalisation cellulaire, UMR 7242 CNRS, Université de Strasbourg, 300 Boulevard Sébastien Brant CS 10413, Illkirch cedex, 67412, France
| | - Nwife Getrude Okechukwu
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
| | - Christian Klein
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
| | - Christine Patte-Mensah
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
| | - Ayikoe-Guy Mensah-Nyagan
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
| |
Collapse
|
17
|
Fu N, Wang Y, Zhu R, Li N, Zeng S, Miao M, Yang Y, Sun M, Zhang J. Bicuculline and Bumetanide Attenuate Sevoflurane-Induced Impairment of Myelination and Cognition in Young Mice. ACS Chem Neurosci 2023; 14:1146-1155. [PMID: 36802490 DOI: 10.1021/acschemneuro.2c00764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Sevoflurane (Sevo) is one of the most commonly used general anesthetics for infants and young children. We investigated whether Sevo impairs neurological functions, myelination, and cognition via the γ-aminobutyric acid A receptor (GABAAR) and Na+-K+-2Cl- cotransporter (NKCC1) in neonatal mice. On postnatal days 5-7, mice were exposed to 3% Sevo for 2 h. On postnatal day 14, mouse brains were dissected, and oligodendrocyte precursor cell line level lentivirus knockdown of GABRB3, immunofluorescence, and transwell migration assays were performed. Finally, behavioral tests were conducted. Multiple Sevo exposure groups exhibited increased neuronal apoptosis levels and decreased neurofilament protein levels in the mouse cortex compared with the control group. Sevo exposure inhibited the proliferation, differentiation, and migration of the oligodendrocyte precursor cells, thereby affecting their maturation process. Electron microscopy revealed that Sevo exposure reduced myelin sheath thickness. The behavioral tests showed that multiple Sevo exposures induced cognitive impairment. GABAAR and NKCC1 inhibition provided protection against Sevo-induced neurotoxicity and cognitive dysfunction. Thus, bicuculline and bumetanide can protect against Sevo-induced neuronal injury, myelination impairment, and cognitive dysfunction in neonatal mice. Furthermore, GABAAR and NKCC1 may be mediators of Sevo-induced myelination impairment and cognitive dysfunction.
Collapse
Affiliation(s)
- Ningning Fu
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China.,Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yangyang Wang
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Ruilou Zhu
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Ningning Li
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China.,Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Shuang Zeng
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China.,Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Mengrong Miao
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Yitian Yang
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Mingyang Sun
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Jiaqiang Zhang
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
18
|
Chen K, Cambi F, Kozai TDY. Pro-myelinating Clemastine administration improves recording performance of chronically implanted microelectrodes and nearby neuronal health. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.31.526463. [PMID: 36778360 PMCID: PMC9915570 DOI: 10.1101/2023.01.31.526463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Intracortical microelectrodes have become a useful tool in neuroprosthetic applications in the clinic and to understand neurological disorders in basic neurosciences. Many of these brain-machine interface technology applications require successful long-term implantation with high stability and sensitivity. However, the intrinsic tissue reaction caused by implantation remains a major failure mechanism causing loss of recorded signal quality over time. Oligodendrocytes remain an underappreciated intervention target to improve chronic recording performance. These cells can accelerate action potential propagation and provides direct metabolic support for neuronal health and functionality. However, implantation injury causes oligodendrocyte degeneration and leads to progressive demyelination in surrounding brain tissue. Previous work highlighted that healthy oligodendrocytes are necessary for greater electrophysiological recording performance and the prevention of neuronal silencing around implanted microelectrodes over chronic implantation. Thus, we hypothesize that enhancing oligodendrocyte activity with a pharmaceutical drug, Clemastine, will prevent the chronic decline of microelectrode recording performance. Electrophysiological evaluation showed that the promyelination Clemastine treatment significantly elevated the signal detectability and quality, rescued the loss of multi-unit activity, and increased functional interlaminar connectivity over 16-weeks of implantation. Additionally, post-mortem immunohistochemistry showed that increased oligodendrocyte density and myelination coincided with increased survival of both excitatory and inhibitory neurons near the implant. Overall, we showed a positive relationship between enhanced oligodendrocyte activity and neuronal health and functionality near the chronically implanted microelectrode. This study shows that therapeutic strategy that enhance oligodendrocyte activity is effective for integrating the functional device interface with brain tissue over chronic implantation period. Abstract Figure
Collapse
|
19
|
Spindler M, Thiel CM. Hypothalamic microstructure and function are related to body mass, but not mental or cognitive abilities across the adult lifespan. GeroScience 2023; 45:277-291. [PMID: 35896889 PMCID: PMC9886766 DOI: 10.1007/s11357-022-00630-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 07/20/2022] [Indexed: 02/03/2023] Open
Abstract
Physical, mental, and cognitive resources are essential for healthy aging. Aging impacts on the structural integrity of various brain regions, including the hippocampus. Even though recent rodent studies hint towards a critical role of the hypothalamus, there is limited evidence on functional consequences of age-related changes of this region in humans. Given its central role in metabolic regulation and affective processing and its connections to the hippocampus, it is plausible that hypothalamic integrity and connectivity are associated with functional age-related decline. We used data of n = 369 participants (18-88 years) from the Cambridge Centre for Ageing and Neuroscience repository to determine functional impacts of potential changes in hypothalamic microstructure across the lifespan. First, we identified age-related changes in microstructure as a function of physical, mental, and cognitive health and compared those findings to changes in hippocampal microstructure. Second, we investigated the relationship of hypothalamic microstructure and resting-state functional connectivity and related those changes to age as well as physical health. Our results showed that hypothalamic microstructure is not affected by depressive symptoms (mental health), cognitive performance (cognitive health), and comparatively stable across the lifespan, but affected by body mass (physical health). Furthermore, body mass changes connectivity to limbic regions including the hippocampus, amygdala, and nucleus accumbens, suggesting functional alterations in the metabolic and reward systems. Our results demonstrate that hypothalamic structure and function are affected by body mass, focused on neural density and dispersion, but not inflammation. Still, observed effect sizes were small, encouraging detailed investigations of individual hypothalamic subunits.
Collapse
Affiliation(s)
- Melanie Spindler
- Biological Psychology, Department of Psychology, School of Medicine and Health Sciences, Carl Von Ossietzky Universität Oldenburg, 26129, Oldenburg, Germany.
| | - Christiane M Thiel
- Biological Psychology, Department of Psychology, School of Medicine and Health Sciences, Carl Von Ossietzky Universität Oldenburg, 26129, Oldenburg, Germany
- Cluster of Excellence "Hearing4all", Carl Von Ossietzky Universität Oldenburg, 26129, Oldenburg, Germany
- Research Centre Neurosensory Science, Carl Von Ossietzky Universität Oldenburg, 26129, Oldenburg, Germany
| |
Collapse
|
20
|
Granato G, Borghi AM, Mattera A, Baldassarre G. A computational model of inner speech supporting flexible goal-directed behaviour in Autism. Sci Rep 2022; 12:14198. [PMID: 35987942 PMCID: PMC9392752 DOI: 10.1038/s41598-022-18445-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/11/2022] [Indexed: 11/21/2022] Open
Abstract
Experimental and computational studies propose that inner speech boosts categorisation skills and executive functions, making human behaviour more focused and flexible. In addition, many clinical studies highlight a relationship between poor inner-speech and an executive impairment in autism spectrum condition (ASC), but contrasting findings are reported. Here we directly investigate the latter issue through a previously implemented and validated computational model of the Wisconsin Cards Sorting Tests. In particular, the model was applied to explore potential individual differences in cognitive flexibility and inner speech contribution in autistic and neurotypical participants. Our model predicts that the use of inner-speech could increase along the life-span of neurotypical participants but would be reduced in autistic ones. Although we found more attentional failures (i.e., wrong behavioural rule switches) in autistic children/teenagers and more perseverative behaviours in autistic young/older adults, only autistic children and older adults exhibited a lower performance (i.e., fewer consecutive correct rule switches) than matched control groups. Overall, our results corroborate the idea that the reduced use of inner speech could represent a disadvantage for autistic children and autistic older adults. Moreover, the results suggest that cognitive-behavioural therapies should focus on developing inner speech skills in autistic children as this could provide cognitive support throughout their whole life span.
Collapse
Affiliation(s)
- Giovanni Granato
- Laboratory of Computational Embodied Neuroscience, Institute of Cognitive Sciences and Technologies, National Research Council of Italy, Rome, Italy.
- School of Computing, Electronics and Mathematics, University of Plymouth, Plymouth, UK.
| | - Anna M Borghi
- Dipartimento di Psicologia Dinamica, Clinica e Salute, Sapienza University of Rome, Institute of Cognitive Sciences and Technologies, National Research Council of Italy, Rome, Italy
| | - Andrea Mattera
- Laboratory of Computational Embodied Neuroscience, Institute of Cognitive Sciences and Technologies, National Research Council of Italy, Rome, Italy
| | - Gianluca Baldassarre
- Laboratory of Computational Embodied Neuroscience, Institute of Cognitive Sciences and Technologies, National Research Council of Italy, Rome, Italy
| |
Collapse
|
21
|
Tsivelekas KK, Evangelopoulos DS, Pallis D, Benetos IS, Papadakis SA, Vlamis J, Pneumaticos SG. Angiogenesis in Spinal Cord Injury: Progress and Treatment. Cureus 2022; 14:e25475. [PMID: 35800787 PMCID: PMC9246426 DOI: 10.7759/cureus.25475] [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] [Accepted: 05/29/2022] [Indexed: 11/22/2022] Open
Abstract
Traumatic spinal cord injury (SCI) provokes the onset of an intricate pathological process. Initial primary injury ruptures local micro-neuro-vascularcomplex triggering the commencement of multi-factorial secondary sequences which exert significant influence on neurological deterioration progress. Stimulating by local ischemia, neovascularization pathways emerge to provide neuroprotection and improve functional recovery. Although angiogenetic processes are prompted, newly formed vascular system is frequently inadequate to distribute sufficient blood supply and improve axonal recovery. Several treatment interventions have been endeavored to achieve the optimal conditions in SCI microenvironment, enhancing angiogenesis and improve functional recovery. In this study we review the revascularization pathogenesis and importance within the secondary processes and condense the proangiogenic influence of several angiogenetic-targeted treatment interventions.
Collapse
|
22
|
Duffau H. White Matter Tracts and Diffuse Lower-Grade Gliomas: The Pivotal Role of Myelin Plasticity in the Tumor Pathogenesis, Infiltration Patterns, Functional Consequences and Therapeutic Management. Front Oncol 2022; 12:855587. [PMID: 35311104 PMCID: PMC8924360 DOI: 10.3389/fonc.2022.855587] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 02/14/2022] [Indexed: 12/18/2022] Open
Abstract
For many decades, interactions between diffuse lower-grade glioma (LGG) and brain connectome were neglected. However, the neoplasm progression is intimately linked to its environment, especially the white matter (WM) tracts and their myelin status. First, while the etiopathogenesis of LGG is unclear, this tumor seems to appear during the adolescence, and it is mostly located within anterior and associative cerebral areas. Because these structures correspond to those which were myelinated later in the brain maturation process, WM myelination could play a role in the development of LGG. Second, WM fibers and the myelin characteristics also participate in LGG diffusion, since glioma cells migrate along the subcortical pathways, especially when exhibiting a demyelinated phenotype, which may result in a large invasion of the parenchyma. Third, such a migratory pattern can induce functional (neurological, cognitive and behavioral) disturbances, because myelinated WM tracts represent the main limitation of neuroplastic potential. These parameters are critical for tailoring an individualized therapeutic strategy, both (i) regarding the timing of active treatment(s) which must be proposed earlier, before a too wide glioma infiltration along the WM bundles, (ii) and regarding the anatomic extent of surgical resection and irradiation, which should take account of the subcortical connectivity. Therefore, the new science of connectomics must be integrated in LGG management, based upon an improved understanding of the interplay across glioma dissemination within WM and reactional neural networks reconfiguration, in order to optimize long-term oncological and functional outcomes. To this end, mechanisms of activity-dependent myelin plasticity should be better investigated.
Collapse
Affiliation(s)
- Hugues Duffau
- Department of Neurosurgery, Gui de Chauliac Hospital, Montpellier University Medical Center, Montpellier, France.,Team "Plasticity of Central Nervous System, Stem Cells and Glial Tumors", Institute of Functional Genomics, National Institute for Health and Medical Research (INSERM) U1191, University of Montpellier, Montpellier, France
| |
Collapse
|
23
|
Biochanin A Improves Memory Decline and Brain Pathology in Cuprizone-Induced Mouse Model of Multiple Sclerosis. Behav Sci (Basel) 2022; 12:bs12030070. [PMID: 35323389 PMCID: PMC8945046 DOI: 10.3390/bs12030070] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 12/30/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and neurodegenerative disease of the central nervous system characterized by the demyelination of nerves, neural degeneration, and axonal loss. Cognitive impairment, including memory decline, is a significant feature in MS affecting up to 70% of patients. Thereby, it substantially impacts patients’ quality of life. Biochanin A (BCA) is an o-methylated isoflavone with a wide variety of pharmacological activities, including antioxidant, anti-inflammatory, and neuroprotective activities. Thus, this study aimed to investigate the possible protective effects of BCA on memory decline in the cuprizone (CPZ) model of MS. Thirty Swiss albino male mice (SWR/J) were randomly divided into three groups (n = 10): control (normal chow + i.p. 1:9 mixture of DMSO and PBS), CPZ (0.2% w/w of CPZ mixed into chow + i.p. 1:9 mixture of DMSO and PBS), and CPZ + BCA (0.2% w/w of CPZ mixed into chow + i.p. 40 mg/kg of BCA). At the last week of the study (week 5), a series of behavioral tasks were performed. A grip strength test was performed to assess muscle weakness while Y-maze, novel object recognition task (NORT), and novel arm discrimination task (NADT) were performed to assess memory. Additionally, histological examination of the hippocampus and the prefrontal cortex (PFC) were conducted. BCA administration caused a significant increase in the grip strength compared with the CPZ group. Additionally, BCA significantly improved the mice’s spatial memory in the Y-maze and recognition memory in the NORT and the NADT compared with the CPZ group. Moreover, BCA mitigated neuronal damage in the PFC and the hippocampus after five weeks of administration. In conclusion, our data demonstrates the possible protective effect of BCA against memory deterioration in mice fed with CPZ for five weeks.
Collapse
|
24
|
Allen AT, Heaton EC, Shapiro LP, Butkovich LM, Yount ST, Davies RA, Li DC, Swanson AM, Gourley SL. Inter-individual variability amplified through breeding reveals control of reward-related action strategies by Melanocortin-4 Receptor in the dorsomedial striatum. Commun Biol 2022; 5:116. [PMID: 35136204 PMCID: PMC8825839 DOI: 10.1038/s42003-022-03043-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/06/2022] [Indexed: 12/15/2022] Open
Abstract
In day-to-day life, we often must choose between pursuing familiar behaviors or adjusting behaviors when new strategies might be more fruitful. The dorsomedial striatum (DMS) is indispensable for arbitrating between old and new action strategies. To uncover molecular mechanisms, we trained mice to generate nose poke responses for food, then uncoupled the predictive relationship between one action and its outcome. We then bred the mice that failed to rapidly modify responding. This breeding created offspring with the same tendencies, failing to inhibit behaviors that were not reinforced. These mice had less post-synaptic density protein 95 in the DMS. Also, densities of the melanocortin-4 receptor (MC4R), a high-affinity receptor for α-melanocyte-stimulating hormone, predicted individuals' response strategies. Specifically, high MC4R levels were associated with poor response inhibition. We next found that reducing Mc4r in the DMS in otherwise typical mice expedited response inhibition, allowing mice to modify behavior when rewards were unavailable or lost value. This process required inputs from the orbitofrontal cortex, a brain region canonically associated with response strategy switching. Thus, MC4R in the DMS appears to propel reward-seeking behavior, even when it is not fruitful, while moderating MC4R presence increases the capacity of mice to inhibit such behaviors.
Collapse
Affiliation(s)
- Aylet T Allen
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Elizabeth C Heaton
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Graduate Program in Neuroscience, Emory University, Atlanta, GA, USA
| | - Lauren P Shapiro
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Graduate Program in Molecular and Systems Pharmacology, Emory University, Atlanta, GA, USA
| | - Laura M Butkovich
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Sophie T Yount
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Graduate Program in Molecular and Systems Pharmacology, Emory University, Atlanta, GA, USA
| | - Rachel A Davies
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Dan C Li
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Graduate Program in Neuroscience, Emory University, Atlanta, GA, USA
| | - Andrew M Swanson
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Graduate Program in Neuroscience, Emory University, Atlanta, GA, USA
| | - Shannon L Gourley
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, USA.
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.
- Graduate Program in Neuroscience, Emory University, Atlanta, GA, USA.
- Graduate Program in Molecular and Systems Pharmacology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
25
|
Leyrolle Q, Decoeur F, Dejean C, Brière G, Leon S, Bakoyiannis I, Baroux E, Sterley TL, Bosch-Bouju C, Morel L, Amadieu C, Lecours C, St-Pierre MK, Bordeleau M, De Smedt-Peyrusse V, Séré A, Schwendimann L, Grégoire S, Bretillon L, Acar N, Joffre C, Ferreira G, Uricaru R, Thebault P, Gressens P, Tremblay ME, Layé S, Nadjar A. N-3 PUFA deficiency disrupts oligodendrocyte maturation and myelin integrity during brain development. Glia 2022; 70:50-70. [PMID: 34519378 DOI: 10.1002/glia.24088] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 12/14/2022]
Abstract
Westernization of dietary habits has led to a progressive reduction in dietary intake of n-3 polyunsaturated fatty acids (n-3 PUFAs). Low maternal intake of n-3 PUFAs has been linked to neurodevelopmental disorders, conditions in which myelination processes are abnormal, leading to defects in brain functional connectivity. Only little is known about the role of n-3 PUFAs in oligodendrocyte physiology and white matter development. Here, we show that lifelong n-3 PUFA deficiency disrupts oligodendrocytes maturation and myelination processes during the postnatal period in mice. This has long-term deleterious consequences on white matter organization and hippocampus-prefrontal functional connectivity in adults, associated with cognitive and emotional disorders. Promoting developmental myelination with clemastine, a first-generation histamine antagonist and enhancer of oligodendrocyte precursor cell differentiation, rescues memory deficits in n-3 PUFA deficient animals. Our findings identify a novel mechanism through which n-3 PUFA deficiency alters brain functions by disrupting oligodendrocyte maturation and brain myelination during the neurodevelopmental period.
Collapse
Affiliation(s)
- Quentin Leyrolle
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France.,Université de Paris, NeuroDiderot, Inserm, Paris, France
| | - Fanny Decoeur
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Cyril Dejean
- Université de Bordeaux, INSERM, Magendie, U1215, F-3300, Bordeaux, France
| | | | - Stephane Leon
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | | | - Emilie Baroux
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Tony-Lee Sterley
- Hotchkiss Brain Institute and the Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | | | - Lydie Morel
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Camille Amadieu
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Cynthia Lecours
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, Québec, Canada.,Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Marie-Kim St-Pierre
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, Québec, Canada.,Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Maude Bordeleau
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, Québec, Canada.,Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada.,Integrated Program in Neuroscience, McGill University, Montréal, Québec City, Québec, Canada
| | | | - Alexandran Séré
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | | | - Stephane Grégoire
- Eye and Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | - Lionel Bretillon
- Eye and Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | - Niyazi Acar
- Eye and Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | - Corinne Joffre
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Guillaume Ferreira
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Raluca Uricaru
- CNRS, Bordeaux INP, LaBRI, UMR 5800, F-33400, Talence, France
| | | | | | - Marie-Eve Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, Québec, Canada.,Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada.,Neurology and Neurosurgery Department, McGill University, Montreal, Québec City, Québec, Canada.,Department of Molecular Medicine, Université Laval, Québec City, Québec, Canada.,Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Sophie Layé
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Agnes Nadjar
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France.,Université de Bordeaux, INSERM, Magendie, U1215, F-3300, Bordeaux, France.,Institut Universitaire de France, Paris, France
| |
Collapse
|
26
|
Abstract
The pathology of fetal alcohol syndrome and the less severe fetal alcohol spectrum disorders includes brain dysmyelination. Recent studies have shed light on the molecular mechanisms underlying these white matter abnormalities. Rodent models of fetal alcohol syndrome and human studies have shown suppressed oligodendrocyte differentiation and apoptosis of oligodendrocyte precursor cells. Ethanol exposure led to reduced expression of myelin basic protein and delayed myelin basic protein expression in rat and mouse models of fetal alcohol syndrome and in human histopathological specimens. Several studies have reported increased expression of many chemokines in dysmyelinating disorders in central nervous system, including multiple sclerosis and fetal alcohol syndrome. Acute ethanol exposure reduced levels of the neuroprotective insulin-like growth factor-1 in fetal and maternal sheep and in human fetal brain tissues, while ethanol increased the expression of tumor necrosis factor α in mouse and human neurons. White matter lesions have been induced in the developing sheep brain by alcohol exposure in early gestation. Rat fetal alcohol syndrome models have shown reduced axon diameters, with thinner myelin sheaths, as well as reduced numbers of oligodendrocytes, which were also morphologically aberrant oligodendrocytes. Expressions of markers for mature myelination, including myelin basic protein, also were reduced. The accumulating knowledge concerning the mechanisms of ethanol-induced dysmyelination could lead to the development of strategies to prevent dysmyelination in children exposed to ethanol during fetal development. Future studies using fetal oligodendrocyte- and oligodendrocyte precursor cell-derived exosomes isolated from the mother’s blood may identify biomarkers for fetal alcohol syndrome and even implicate epigenetic changes in early development that affect oligodendrocyte precursor cell and oligodendrocyte function in adulthood. By combining various imaging modalities with molecular studies, it may be possible to determine which fetuses are at risk and to intervene therapeutically early in the pregnancy.
Collapse
Affiliation(s)
- Nune Darbinian
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Michael E Selzer
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
27
|
Lynch CMK, Nagpal J, Clarke G, Cryan JF. Wrapping Things Up: Recent Developments in Understanding the Role of the Microbiome in Regulating Myelination. CURRENT OPINION IN PHYSIOLOGY 2021. [DOI: 10.1016/j.cophys.2021.100468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
28
|
Silva-Gotay A, Davis J, Tavares ER, Richardson HN. Alcohol drinking during early adolescence activates microglial cells and increases frontolimbic Interleukin-1 beta and Toll-like receptor 4 gene expression, with heightened sensitivity in male rats compared to females. Neuropharmacology 2021; 197:108698. [PMID: 34252404 PMCID: PMC8552486 DOI: 10.1016/j.neuropharm.2021.108698] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 05/13/2021] [Accepted: 06/15/2021] [Indexed: 12/31/2022]
Abstract
Adolescent drinking is risky because neural circuits in the frontal lobes are undergoing maturational processes important for cognitive function and behavioral control in adulthood. Previous studies have shown that myelinated axons in the medial prefrontal cortex (mPFC) are particularly sensitive to alcohol drinking, especially in males. Pro-inflammatory mediators like toll-like receptor 4 (TLR4) and interleukin-1 beta (IL1b) have been implicated in alcohol induced-inflammation and demyelination; thus, herein we test the hypothesis that voluntary alcohol drinking early in adolescence elicits a pro-inflammatory state that is more pronounced in the brain of males compared to females. Adolescent male and female Wistar rats self-administered sweetened alcohol or sweetened water from postnatal days 28-42 and separate sets of brains were processed for 1) immunolabeling for ionized calcium-binding adapter molecule 1 to analyze microglial cell morphology, or 2) qPCR analysis of gene expression of pro-inflammatory mediators. Binge drinking alcohol activated microglia in the mPFC and hippocampus of both males and females, suggesting that voluntary alcohol exposure initiates an inflammatory response. Il1b mRNA was upregulated in the mPFC of both sexes. Conversely, Tlr4 mRNA levels were elevated after drinking only in males, which could explain more robust effects of alcohol on myelin in this region in developing males compared to females. Il1b mRNA changes were not observed in the hippocampus, but alcohol elevated Tlr4 mRNA in both sexes, highlighting regional specificity in inflammatory responses to alcohol. Overall, these findings give insight into potential mechanisms by which low-to-moderate voluntary alcohol intake impacts the developing brain. This article is part of the special Issue on 'Vulnerabilities to Substance Abuse'.
Collapse
Affiliation(s)
- Andrea Silva-Gotay
- Neuroscience and Behavior Graduate Program, University of
Massachusetts, Amherst, MA 01003
| | - Jillian Davis
- Department of Psychological and Brain Sciences, University
of Massachusetts, Amherst, MA 01003
| | - Elizabeth R. Tavares
- Department of Psychological and Brain Sciences, University
of Massachusetts, Amherst, MA 01003
| | - Heather N. Richardson
- Neuroscience and Behavior Graduate Program, University of
Massachusetts, Amherst, MA 01003,Department of Psychological and Brain Sciences, University
of Massachusetts, Amherst, MA 01003
| |
Collapse
|
29
|
Wang Z, Baharani A, Wei Z, Truong D, Bi X, Wang F, Li XM, Verge VMK, Zhang Y. Low field magnetic stimulation promotes myelin repair and cognitive recovery in chronic cuprizone mouse model. Clin Exp Pharmacol Physiol 2021; 48:1090-1102. [PMID: 33638234 DOI: 10.1111/1440-1681.13490] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/09/2021] [Accepted: 02/23/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is an inflammatory demyelinating disease featured with neuroinflammation, demyelination, and the loss of oligodendrocytes. Cognitive impairment and depression are common neuropsychiatric symptoms in MS that are poorly managed with the present interventions. OBJECTIVE This study aimed to investigate the effects of low field magnetic stimulation (LFMS), a novel non-invasive neuromodulation technology, on cognitive impairment and depressive symptoms associated with MS using a mouse model of demyelination. METHODS C57BL female mice were fed with a 0.2% cuprizone diet for 12 weeks to induce a chronic demyelinating model followed by 4 weeks of cuprizone withdrawal with either sham or LFMS treatment. RESULTS Improved cognition and depression-like behaviour and restored weight gain were observed in mice with LFMS treatment. Immunohistochemical and immunoblotting data showed enhanced myelin basic protein (MBP) and myelin oligodendrocyte glycoprotein expressions (MOG) in the prefrontal cortex of mice with LFMS treatment, supporting that myelin repair was promoted. LFMS also increased the protein expression of mature oligodendrocyte biomarker glutathione-S-transferase (GST-π). In addition, expression of TGF-β and associated receptors were elevated with LFMS treatment, implicating this pathway in the response. CONCLUSION Results from the present study revealed LFMS to have neuroprotective effects, suggesting that LFMS has potential therapeutic value for treating cognitive impairment and depression related to demyelination disorders.
Collapse
Affiliation(s)
- Zitong Wang
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Akanksha Baharani
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Zelan Wei
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Davin Truong
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiaoying Bi
- Department of Neurology, Shanghai Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Fei Wang
- Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Xin-Min Li
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Valerie M K Verge
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
- Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Yanbo Zhang
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
- Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
30
|
Zhang Y, Al Mamun A, Yuan Y, Lu Q, Xiong J, Yang S, Wu C, Wu Y, Wang J. Acute spinal cord injury: Pathophysiology and pharmacological intervention (Review). Mol Med Rep 2021; 23:417. [PMID: 33846780 PMCID: PMC8025476 DOI: 10.3892/mmr.2021.12056] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injury (SCI) is one of the most debilitating of all the traumatic conditions that afflict individuals. For a number of years, extensive studies have been conducted to clarify the molecular mechanisms of SCI. Experimental and clinical studies have indicated that two phases, primary damage and secondary damage, are involved in SCI. The initial mechanical damage is caused by local impairment of the spinal cord. In addition, the fundamental mechanisms are associated with hyperflexion, hyperextension, axial loading and rotation. By contrast, secondary injury mechanisms are led by systemic and cellular factors, which may also be initiated by the primary injury. Although significant advances in supportive care have improved clinical outcomes in recent years, a number of studies continue to explore specific pharmacological therapies to minimize SCI. The present review summarized some important pathophysiologic mechanisms that are involved in SCI and focused on several pharmacological and non‑pharmacological therapies, which have either been previously investigated or have a potential in the management of this debilitating injury in the near future.
Collapse
Affiliation(s)
- Yi Zhang
- School of Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu 210094, P.R. China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Abdullah Al Mamun
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Yuan Yuan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Qi Lu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Jun Xiong
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Shulin Yang
- School of Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu 210094, P.R. China
| | - Chengbiao Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Yanqing Wu
- Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, P.R. China
| | - Jian Wang
- Department of Hand Surgery and Peripheral Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| |
Collapse
|
31
|
Carolien K, Annika M. Affective decision-making in children prenatally exposed to opioids. Scand J Psychol 2021; 62:529-536. [PMID: 34037260 DOI: 10.1111/sjop.12743] [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: 11/19/2020] [Accepted: 04/01/2021] [Indexed: 11/29/2022]
Abstract
Although opioid maintenance therapy (OMT) is currently recommended for pregnant opioid-dependent women, potential effects on children's long-term development are still largely unknown. The current study assessed the long-term cognitive development of children born to women in OMT. Particularly, children's decision-making performance was assessed with a child-friendly version of the Iowa Gambling Task. Using a prospective longitudinal design, a cohort of children was followed from birth to middle childhood. Data were collected in Norway between 2005 and 2017. Participants included 41 children (aged 9-11 years), 20 of whom had histories of prenatal methadone or buprenorphine exposure. Background data were collected from personal interviews and medical records in 2005-2006. Children's affective decision-making was assessed in 2016-2017. Results showed no main effect of group on the net scores in the gambling task, F(1, 39) = 1.44, p = 0.24, η2 = 0.04, demonstrating no group differences in decision-making performance. A main effect of group was found on sensitivity to punishment, with children in the control group choosing the doors with the infrequent, but high punishment more often compared to children in the OMT group, F(1, 39) = 4.90, p = 0.03, η2 = 0.11. No main effect of group on decision-making speed was found, although results showed a significant interaction effect between group and gain, F(1, 8,194) = 4.09, p = 0.04, η2 = 0.001. Children prenatally exposed to opioids were found to have normal decision-making performance on an affective decision-making task and were able to consider future consequences when making decisions.
Collapse
Affiliation(s)
- Konijnenberg Carolien
- Department of Psychology, Inland Norway University of Applied Sciences, Lillehammer, Norway.,Cognitive Developmental Research Unit, Department of Psychology, University of Oslo, Oslo, Norway
| | - Melinder Annika
- Cognitive Developmental Research Unit, Department of Psychology, University of Oslo, Oslo, Norway.,Child- and Adolescents Mental Health, Oslo University hospital, Oslo, Norway
| |
Collapse
|
32
|
Lin Q, Wang S, Duan Y, Tuchin VV. Ex vivo three-dimensional elemental imaging of mouse brain tissue block by laser-induced breakdown spectroscopy. JOURNAL OF BIOPHOTONICS 2021; 14:e202000479. [PMID: 33512064 DOI: 10.1002/jbio.202000479] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 06/12/2023]
Abstract
Measurement and reconstruction of an elemental image of large brain tissue will be beneficial to the diagnosis of neurological brain diseases. Herein, laser-induced breakdown spectroscopy (LIBS) is introduced for three dimensional (3D) elemental analysis of paraffin-embedded mouse brain tissue blocks. It is used for the first time towards the mapping of mouse brain block samples. A micro-LIBS prototype is developed for brain elemental imaging and a layer-by-layer approach is used to reconstruct the 3D distribution of Ca, Mg, Na, Cu, and P in the brain tissue. Images are captured with 50 μm lateral resolution and 300 μm depth resolution. The images show that the reclamation area of the cortex surface is enriched with Ca and Mg. In contrast, the Cu distribution is circular and is found primarily in the entirety of the cerebral cortex for the paraffin-embedded brain samples. Elemental imaging results suggest that the highest P intensity is found in the cerebellum nearby the middle sagittal plane in the left-brain paraffin block. These preliminary results indicate that LIBS is a potentially powerful tool for elemental bioimaging of the whole brain and may further improve the understanding of complex brain mechanisms.
Collapse
Affiliation(s)
- Qingyu Lin
- School of Mechanical Engineering, Research Center of Analytical Instrumentation, Sichuan University, Chengdu, China
| | - Shuai Wang
- Kunming Institute of Physics, Kunming, China
| | - Yixiang Duan
- School of Mechanical Engineering, Research Center of Analytical Instrumentation, Sichuan University, Chengdu, China
| | - Valery V Tuchin
- Research-Educational Institute of Optics and Biophotonics, Saratov State University, Saratov, Russia
- Laboratory of Laser Diagnostics of Technical and Living Systems, Institute of Precision Mechanics and Control of the RAS, Saratov, Russia
- Interdisciplinary Laboratory of Biophotonics, Tomsk State University, Tomsk, Russia
| |
Collapse
|
33
|
Amaya JM, Suidgeest E, Sahut-Barnola I, Dumontet T, Montanier N, Pagès G, Keller C, van der Weerd L, Pereira AM, Martinez A, Meijer OC. Effects of Long-Term Endogenous Corticosteroid Exposure on Brain Volume and Glial Cells in the AdKO Mouse. Front Neurosci 2021; 15:604103. [PMID: 33642975 PMCID: PMC7902940 DOI: 10.3389/fnins.2021.604103] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/06/2021] [Indexed: 01/26/2023] Open
Abstract
Chronic exposure to high circulating levels of glucocorticoids has detrimental effects on health, including metabolic abnormalities, as exemplified in Cushing’s syndrome (CS). Magnetic resonance imaging (MRI) studies have found volumetric changes in gray and white matter of the brain in CS patients during the course of active disease, but also in remission. In order to explore this further, we performed MRI-based brain volumetric analyses in the AdKO mouse model for CS, which presents its key traits. AdKO mice had reduced relative volumes in several brain regions, including the corpus callosum and cortical areas. The medial amygdala, bed nucleus of the stria terminalis, and hypothalamus were increased in relative volume. Furthermore, we found a lower immunoreactivity of myelin basic protein (MBP, an oligodendrocyte marker) in several brain regions but a paradoxically increased MBP signal in the male cingulate cortex. We also observed a decrease in the expression of glial fibrillary acidic protein (GFAP, a marker for reactive astrocytes) and ionized calcium-binding adapter molecule 1 (IBA1, a marker for activated microglia) in the cingulate regions of the anterior corpus callosum and the hippocampus. We conclude that long-term hypercorticosteronemia induced brain region-specific changes that might include aberrant myelination and a degree of white matter damage, as both repair (GFAP) and immune (IBA1) responses are decreased. These findings suggest a cause for the changes observed in the brains of human patients and serve as a background for further exploration of their subcellular and molecular mechanisms.
Collapse
Affiliation(s)
- Jorge Miguel Amaya
- Department of Endocrinology, Leiden University Medical Center, Leiden, Netherlands
| | - Ernst Suidgeest
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Isabelle Sahut-Barnola
- Génétique Reproduction et Développement, Université Clermont-Auvergne, CNRS, INSERM, Clermont-Ferrand, France
| | - Typhanie Dumontet
- Génétique Reproduction et Développement, Université Clermont-Auvergne, CNRS, INSERM, Clermont-Ferrand, France
| | - Nathanaëlle Montanier
- Génétique Reproduction et Développement, Université Clermont-Auvergne, CNRS, INSERM, Clermont-Ferrand, France
| | - Guilhem Pagès
- INRAE, AgroResonance, QuaPA UR370, Saint-Genès-Champanelle, France
| | - Cécile Keller
- INRAE, AgroResonance, QuaPA UR370, Saint-Genès-Champanelle, France
| | - Louise van der Weerd
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands.,Human Genetics Department, Leiden University Medical Center, Leiden, Netherlands
| | - Alberto M Pereira
- Department of Endocrinology, Leiden University Medical Center, Leiden, Netherlands
| | - Antoine Martinez
- INRAE, AgroResonance, QuaPA UR370, Saint-Genès-Champanelle, France
| | - Onno C Meijer
- Department of Endocrinology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
34
|
A gene expression atlas for different kinds of stress in the mouse brain. Sci Data 2020; 7:437. [PMID: 33328476 PMCID: PMC7744580 DOI: 10.1038/s41597-020-00772-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 11/25/2020] [Indexed: 12/17/2022] Open
Abstract
Stressful experiences are part of everyday life and animals have evolved physiological and behavioral responses aimed at coping with stress and maintaining homeostasis. However, repeated or intense stress can induce maladaptive reactions leading to behavioral disorders. Adaptations in the brain, mediated by changes in gene expression, have a crucial role in the stress response. Recent years have seen a tremendous increase in studies on the transcriptional effects of stress. The input raw data are freely available from public repositories and represent a wealth of information for further global and integrative retrospective analyses. We downloaded from the Sequence Read Archive 751 samples (SRA-experiments), from 18 independent BioProjects studying the effects of different stressors on the brain transcriptome in mice. We performed a massive bioinformatics re-analysis applying a single, standardized pipeline for computing differential gene expression. This data mining allowed the identification of novel candidate stress-related genes and specific signatures associated with different stress conditions. The large amount of computational results produced was systematized in the interactive “Stress Mice Portal”.
Collapse
|
35
|
DCX + neuronal progenitors contribute to new oligodendrocytes during remyelination in the hippocampus. Sci Rep 2020; 10:20095. [PMID: 33208869 PMCID: PMC7674453 DOI: 10.1038/s41598-020-77115-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
A pool of different types of neural progenitor cells resides in the adult hippocampus. Apart from doublecortin-expressing (DCX+) neuronal progenitor cells (NPCs), the hippocampal parenchyma also contains oligodendrocyte precursor cells (OPCs), which can differentiate into myelinating oligodendrocytes. It is not clear yet to what extent the functions of these different progenitor cell types overlap and how plastic these cells are in response to pathological processes. The aim of this study was to investigate whether hippocampal DCX+ NPCs can generate new oligodendrocytes under conditions in which myelin repair is required. For this, the cell fate of DCX-expressing NPCs was analyzed during cuprizone-induced demyelination and subsequent remyelination in two regions of the hippocampal dentate gyrus of DCX-CreERT2/Flox-EGFP transgenic mice. In this DCX reporter model, the number of GFP+ NPCs co-expressing Olig2 and CC1, a combination of markers typically found in mature oligodendrocytes, was significantly increased in the hippocampal DG during remyelination. In contrast, the numbers of GFP+PDGFRα+ cells, as well as their proliferation, were unaffected by de- or remyelination. During remyelination, a higher portion of newly generated BrdU-labeled cells were GFP+ NPCs and there was an increase in new oligodendrocytes derived from these proliferating cells (GFP+Olig2+BrdU+). These results suggest that DCX-expressing NPCs were able to contribute to the generation of mature oligodendrocytes during remyelination in the adult hippocampus.
Collapse
|
36
|
Darbinian N, Darbinyan A, Merabova N, Bajwa A, Tatevosian G, Martirosyan D, Zhao H, Selzer ME, Goetzl L. Ethanol-mediated alterations in oligodendrocyte differentiation in the developing brain. Neurobiol Dis 2020; 148:105181. [PMID: 33189883 DOI: 10.1016/j.nbd.2020.105181] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/21/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Alterations of white matter integrity and subsequent white matter structural deficits are consistent findings in Fetal Alcohol Syndrome (FAS), but knowledge regarding the molecular mechanisms underlying these abnormalities is incomplete. Experimental rodent models of FAS have shown dysregulation of cytokine expression leading to apoptosis of oligodendrocyte precursor cells (OPCs) and altered oligodendrocyte (OL) differentiation, but whether this is representative of human FAS pathogenesis has not been determined. METHODS Fetal brain tissue (12.2-21.4 weeks gestation) from subjects undergoing elective termination of pregnancy was collected according to an IRB-approved protocol. Ethanol (EtOH) exposure status was classified based on a detailed face-to-face questionnaire adapted from the National Institute on Alcohol Abuse and Alcoholism Prenatal Alcohol and Sudden Infant Death Syndrome and Stillbirth (PASS) study. Twenty EtOH-exposed fetuses were compared with 20 gestational age matched controls. Cytokine and OPC marker mRNA expression was quantified by Real-Time Polymerase chain reaction (qRT-PCR). Patterns of protein expression of OPC markers and active Capase-3 were studied by Fluorescence Activated Cell Sorting (FACS). RESULTS EtOH exposure was associated with reduced markers of cell viability, OPC differentiation, and OL maturation, while early OL differentiation markers were unchanged or increased. Expression of mRNAs for proteins specific to more mature forms of OL lineage (platelet-derived growth factor α (PDGFRα) and myelin basic protein (MBP) was lower in the EtOH group than in controls. Expression of the multifunctional growth and differentiation-promoting growth factor IGF-1, which is essential for normal development, also was reduced. Reductions were not observed for markers of early stages of OL differentiation, including Nuclear transcription factor NK-2 homeobox locus 2 (Nkx2.2). Expression of mRNAs for the proinflammatory cytokine, tumor necrosis factor-α (TNFα), and several proinflammatory chemokines was higher in the EtOH group compared to controls, including: Growth regulated protein alpha/chemokine (C-X-C motif) ligand 1 (GRO-α/CXCL1), Interleukin 8/chemokine (C-X-C motif) ligand 8 (IL8/CXCL8), Chemokine (C-X-C motif) ligand 6/Granulocyte chemotactic protein 2 (CXCL16/GCP2), epithelial-derived neutrophil-activating protein 78/chemokine (C-X-C motif) ligand 5 (ENA-78/CXCL5), monocyte chemoattractant protein-1 (MCP-1). EtOH exposure also was associated with an increase in the proportion of cells expressing markers of early stage OPCs, such as A2B5 and NG2. Finally, apoptosis (measured by caspase-3 activation) was increased substantially in the EtOH group compared to controls. CONCLUSION Prenatal EtOH exposure is associated with excessive OL apoptosis and/or delayed OL maturation in human fetal brain. This is accompanied by markedly dysregulated expression of several chemokines and cytokines, in a pattern predictive of increased OL cytotoxicity and reduced OL differentiation. These findings are consistent with findings in animal models of FAS.
Collapse
Affiliation(s)
- Nune Darbinian
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States of America.
| | - Armine Darbinyan
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, United States of America.
| | - Nana Merabova
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States of America.
| | - Ahsun Bajwa
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States of America.
| | - Gabriel Tatevosian
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States of America.
| | - Diana Martirosyan
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States of America.
| | - Huaqing Zhao
- Department of Clinical Sciences (Biostatistics and Epidemiology), Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States of America.
| | - Michael E Selzer
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States of America.
| | - Laura Goetzl
- Department of Obstetrics & Gynecology, University of Texas, Houston, TX 77030, United States of America.
| |
Collapse
|
37
|
Kurokawa K, Tsuji M, Takahashi K, Miyagawa K, Mochida-Saito A, Takeda H. Leukemia Inhibitory Factor Participates in the Formation of Stress Adaptation via Hippocampal Myelination in Mice. Neuroscience 2020; 446:1-13. [DOI: 10.1016/j.neuroscience.2020.08.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/27/2022]
|
38
|
De León Reyes NS, Bragg-Gonzalo L, Nieto M. Development and plasticity of the corpus callosum. Development 2020; 147:147/18/dev189738. [PMID: 32988974 DOI: 10.1242/dev.189738] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The corpus callosum (CC) connects the cerebral hemispheres and is the major mammalian commissural tract. It facilitates bilateral sensory integration and higher cognitive functions, and is often affected in neurodevelopmental diseases. Here, we review the mechanisms that contribute to the development of CC circuits in animal models and humans. These species comparisons reveal several commonalities. First, there is an early period of massive axonal projection. Second, there is a postnatal temporal window, varying between species, in which early callosal projections are selectively refined. Third, sensory-derived activity influences axonal refinement. We also discuss how defects in CC formation can lead to mild or severe CC congenital malformations.
Collapse
Affiliation(s)
- Noelia S De León Reyes
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, (CNB-CSIC) Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| | - Lorena Bragg-Gonzalo
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, (CNB-CSIC) Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| | - Marta Nieto
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, (CNB-CSIC) Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| |
Collapse
|
39
|
Casella C, Lipp I, Rosser A, Jones DK, Metzler‐Baddeley C. A Critical Review of White Matter Changes in Huntington's Disease. Mov Disord 2020; 35:1302-1311. [PMID: 32537844 PMCID: PMC9393936 DOI: 10.1002/mds.28109] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/07/2020] [Accepted: 04/30/2020] [Indexed: 12/20/2022] Open
Abstract
Huntington's disease is a genetic neurodegenerative disorder. White matter alterations have recently been identified as a relevant pathophysiological feature of Huntington's disease, but their etiology and role in disease pathogenesis and progression remain unclear. Increasing evidence suggests that white matter changes in this disorder are attributed to alterations in myelin-associated biological processes. This review first discusses evidence from neurochemical studies lending support to the demyelination hypothesis of Huntington's disease, demonstrating aberrant myelination and changes in oligodendrocytes in the Huntington's brain. Next, evidence from neuroimaging studies is reviewed, the limitations of the described methodologies are discussed, and suggested interpretations of findings from published studies are challenged. Although our understanding of Huntington's associated pathological changes in the brain will increasingly rely on neuroimaging techniques, the shortcomings of these methodologies must not be forgotten. Advances in magnetic resonance imaging techniques and tissue modeling will enable a better in vivo, longitudinal characterization of the biological properties of white matter microstructure. This in turn will facilitate identification of disease-related biomarkers and the specification of outcome measures in clinical trials. © 2020 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Chiara Casella
- Cardiff University Brain Research Imaging CentreSchool of Psychology, Cardiff UniversityCardiffUnited Kingdom
| | - Ilona Lipp
- Department of NeurophysicsMax Planck Institute for Human Cognitive and Brain SciencesLeipzigGermany
| | - Anne Rosser
- School of BiosciencesCardiff UniversityCardiffUnited Kingdom
| | - Derek K Jones
- Cardiff University Brain Research Imaging CentreSchool of Psychology, Cardiff UniversityCardiffUnited Kingdom
- Mary MacKillop Institute for Health ResearchAustralian Catholic UniversityMelbourneVictoriaAustralia
| | - Claudia Metzler‐Baddeley
- Cardiff University Brain Research Imaging CentreSchool of Psychology, Cardiff UniversityCardiffUnited Kingdom
| |
Collapse
|
40
|
Interneuron hypomyelination is associated with cognitive inflexibility in a rat model of schizophrenia. Nat Commun 2020; 11:2329. [PMID: 32393757 PMCID: PMC7214427 DOI: 10.1038/s41467-020-16218-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 04/21/2020] [Indexed: 01/13/2023] Open
Abstract
Impaired cognitive functioning is a core feature of schizophrenia, and is hypothesized to be due to myelination as well as interneuron defects during adolescent prefrontal cortex (PFC) development. Here we report that in the apomorphine-susceptible (APO-SUS) rat model, which has schizophrenia-like features, a myelination defect occurred specifically in parvalbumin interneurons. The adult rats displayed medial PFC (mPFC)-dependent cognitive inflexibility, and a reduced number of mature oligodendrocytes and myelinated parvalbumin inhibitory axons in the mPFC. In the developing mPFC, we observed decreased myelin-related gene expression that persisted into adulthood. Environmental enrichment applied during adolescence restored parvalbumin interneuron hypomyelination as well as cognitive inflexibility. Collectively, these findings highlight that impairment of parvalbumin interneuron myelination is related to schizophrenia-relevant cognitive deficits. Dysfunction of GABAergic neurons in the prefrontal cortex has been reported in schizophrenia. Here, the authors use the apomorphine-susceptible rat, which displays some schizophrenia-like behaviors, and show that interneurons in the medial prefrontal cortex are hypomyelinated, which may contribute to this behavioral phenotype.
Collapse
|
41
|
Ferreira BK, Rodrigues MT, Streck EL, Ferreira GC, Schuck PF. White matter disturbances in phenylketonuria: Possible underlying mechanisms. J Neurosci Res 2020; 99:349-360. [PMID: 32141105 DOI: 10.1002/jnr.24598] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/09/2020] [Accepted: 02/04/2020] [Indexed: 12/24/2022]
Abstract
White matter pathologies, as well as intellectual disability, microcephaly, and other central nervous system injuries, are clinical traits commonly ascribed to classic phenylketonuria (PKU). PKU is an inherited metabolic disease elicited by the deficiency of phenylalanine hydroxylase. Accumulation of l-phenylalanine (Phe) and its metabolites is found in tissues and body fluids in phenylketonuric patients. In order to mitigate the clinical findings, rigorous dietary Phe restriction constitutes the core of therapeutic management in PKU. Myelination is the process whereby the oligodendrocytes wrap myelin sheaths around the axons, supporting the conduction of action potentials. White matter injuries are implicated in the brain damage related to PKU, especially in untreated or poorly treated patients. The present review summarizes evidence toward putative mechanisms driving the white matter pathology in PKU patients.
Collapse
Affiliation(s)
- Bruna Klippel Ferreira
- Laboratório de Neuroenergética e Erros Inatos do Metabolismo, Programa de Bioquímica e Biofísica Celular, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Porto Alegre, Brazil
| | - Melissa Torres Rodrigues
- Laboratório de Erros Inatos do Metabolismo, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Programa de Pós-graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Emilio Luiz Streck
- Laboratório de Neurologia Experimental, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Gustavo Costa Ferreira
- Laboratório de Neuroenergética e Erros Inatos do Metabolismo, Programa de Bioquímica e Biofísica Celular, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Porto Alegre, Brazil
| | - Patricia Fernanda Schuck
- Laboratório de Erros Inatos do Metabolismo, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Programa de Pós-graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
42
|
Aksenov DP, Miller MJ, Dixon CJ, Drobyshevsky A. Impact of anesthesia exposure in early development on learning and sensory functions. Dev Psychobiol 2020; 62:559-572. [PMID: 32115695 DOI: 10.1002/dev.21963] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 01/27/2020] [Accepted: 02/08/2020] [Indexed: 12/11/2022]
Abstract
Each year, millions of children undergo anesthesia, and both human and animal studies have indicated that exposure to anesthesia at an early age can lead to neuronal damage and learning deficiency. However, disorders of sensory functions were not reported in children or animals exposed to anesthesia during infancy, which is surprising, given the significant amount of damage to brain tissue reported in many animal studies. In this review, we discuss the relationship between the systems in the brain that mediate sensory input, spatial learning, and classical conditioning, and how these systems could be affected during anesthesia exposure. Based on previous reports, we conclude that anesthesia can induce structural, functional, and compensatory changes in both sensory and learning systems. Changes in myelination following anesthesia exposure were observed as well as the neurodegeneration in the gray matter across variety of brain regions. Disproportionate cell death between excitatory and inhibitory cells induced by anesthesia exposure can lead to a long-term shift in the excitatory/inhibitory balance, which affects both learning-specific networks and sensory systems. Anesthesia may directly affect synaptic plasticity which is especially critical to learning acquisition. However, sensory systems appear to have better ability to compensate for damage than learning-specific networks.
Collapse
Affiliation(s)
| | | | - Conor J Dixon
- NorthShore University HealthSystem, Evanston, IL, USA
| | | |
Collapse
|
43
|
Langnes E, Sneve MH, Sederevicius D, Amlien IK, Walhovd KB, Fjell AM. Anterior and posterior hippocampus macro‐ and microstructure across the lifespan in relation to memory—A longitudinal study. Hippocampus 2020; 30:678-692. [DOI: 10.1002/hipo.23189] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/16/2019] [Accepted: 12/20/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Espen Langnes
- Center for Lifespan Changes in Brain and CognitionUniversity of Oslo Norway
| | - Markus H. Sneve
- Center for Lifespan Changes in Brain and CognitionUniversity of Oslo Norway
| | | | - Inge K. Amlien
- Center for Lifespan Changes in Brain and CognitionUniversity of Oslo Norway
| | - Kristine B. Walhovd
- Center for Lifespan Changes in Brain and CognitionUniversity of Oslo Norway
- Department of Radiology and Nuclear MedicineOslo University Hospital Oslo Norway
| | - Anders M. Fjell
- Center for Lifespan Changes in Brain and CognitionUniversity of Oslo Norway
- Department of Radiology and Nuclear MedicineOslo University Hospital Oslo Norway
| |
Collapse
|
44
|
Boda E. Myelin and oligodendrocyte lineage cell dysfunctions: New players in the etiology and treatment of depression and stress‐related disorders. Eur J Neurosci 2019; 53:281-297. [DOI: 10.1111/ejn.14621] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/06/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Enrica Boda
- Department of Neuroscience Rita Levi‐Montalcini University of Turin Turin Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO) University of Turin Turin Italy
| |
Collapse
|
45
|
Sorond FA, Whitehead S, Arai K, Arnold D, Carmichael ST, De Carli C, Duering M, Fornage M, Flores-Obando RE, Graff-Radford J, Hamel E, Hess DC, Ihara M, Jensen MK, Markus HS, Montagne A, Rosenberg G, Shih AY, Smith EE, Thiel A, Tse KH, Wilcock D, Barone F. Proceedings from the Albert Charitable Trust Inaugural Workshop on white matter and cognition in aging. GeroScience 2019; 42:81-96. [PMID: 31811528 DOI: 10.1007/s11357-019-00141-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 11/20/2019] [Indexed: 12/13/2022] Open
Abstract
This third in a series of vascular cognitive impairment (VCI) workshops, supported by "The Leo and Anne Albert Charitable Trust," was held from February 8 to 12 at the Omni Resort in Carlsbad, CA. This workshop followed the information gathered from the earlier two workshops suggesting that we focus more specifically on brain white matter in age-related cognitive impairment. The Scientific Program Committee (Frank Barone, Shawn Whitehead, Eric Smith, and Rod Corriveau) assembled translational, clinical, and basic scientists with unique expertise in acute and chronic white matter injury at the intersection of cerebrovascular and neurodegenerative etiologies. As in previous Albert Trust workshops, invited participants addressed key topics related to mechanisms of white matter injury, biomarkers of white matter injury, and interventions to prevent white matter injury and age-related cognitive decline. This report provides a synopsis of the presentations and discussions by the participants, including the existing knowledge gaps and the delineation of the next steps towards advancing our understanding of white matter injury and age-related cognitive decline. Workshop discussions and consensus resulted in action by The Albert Trust to (1) increase support from biannual to annual "White Matter and Cognition" workshops; (2) provide funding for two collaborative, novel research grants annually submitted by meeting participants; and (3) coordinate the formation of the "Albert Research Institute for White Matter and Cognition." This institute will fill a gap in white matter science, providing white matter and cognition communications, including annual updates from workshops and the literature and interconnecting with other Albert Trust scientific endeavors in cognition and dementia, and providing support for newly established collaborations between seasoned investigators and to the development of talented young investigators in the VCI-dementia (VCID) and white matter cognition arena.
Collapse
Affiliation(s)
- Farzaneh A Sorond
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA.
| | - Shawn Whitehead
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Ken Arai
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Douglas Arnold
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - S Thomas Carmichael
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Charles De Carli
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Marco Duering
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Myriam Fornage
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Rafael E Flores-Obando
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Jonathan Graff-Radford
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Edith Hamel
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - David C Hess
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Massafumi Ihara
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Majken K Jensen
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Hugh S Markus
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Axel Montagne
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Gary Rosenberg
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Andy Y Shih
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Eric E Smith
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Alex Thiel
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Kai Hei Tse
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Donna Wilcock
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| | - Frank Barone
- Department of Neurology, Division Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, 625 N. Michigan Ave, suite 1150, Chicago, IL, 60611, USA
| |
Collapse
|
46
|
Rice J, Coutellier L, Weiner JL, Gu C. Region-specific interneuron demyelination and heightened anxiety-like behavior induced by adolescent binge alcohol treatment. Acta Neuropathol Commun 2019; 7:173. [PMID: 31703603 DOI: 10.1186/s40478-019-0829-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/13/2019] [Indexed: 12/12/2022] Open
Abstract
Adolescent binge drinking represents a major public health challenge and can lead to persistent neurological and mental conditions, but the underlying pathogenic mechanisms remain poorly understood. Using a mouse model of adolescent binge ethanol treatment (ABET), we found that this treatment induced behavioral changes associated with demyelination in different brain regions. After ABET, adolescent mice exhibited anxiogenic behaviors with no change in locomotion on the elevated plus maze, and impaired spatial memory indicated by a significant reduction in spontaneous alternation in the Y maze test. Both effects persisted into adulthood. Anatomical studies further showed that ABET induced a significant reduction of parvalbumin-positive (PV+) GABAergic interneurons and myelin density in the hippocampus and medial prefrontal cortex (mPFC). While these deficits in PV+ interneurons and myelin persisted into early adulthood in the hippocampus, the myelin density recovered in the mPFC. Moreover, whereas ABET mainly damaged myelin of PV+ axons in the hippocampus, it primarily damaged myelin of PV-negative axons in the mPFC. Thus, our findings reveal that an adolescent binge alcohol treatment regimen disrupts spatial working memory, increases anxiety-like behaviors, and exerts unique temporal and spatial patterns of gray matter demyelination in the hippocampus and mPFC.
Collapse
|
47
|
Early Postnatal Exposure to Isoflurane Disrupts Oligodendrocyte Development and Myelin Formation in the Mouse Hippocampus. Anesthesiology 2019; 131:1077-1091. [PMID: 31436548 PMCID: PMC6800770 DOI: 10.1097/aln.0000000000002904] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Early postnatal exposure to general anesthetics may interfere with brain development. We tested the hypothesis that isoflurane causes a lasting disruption in myelin development via actions on the mammalian target of rapamycin pathway. METHODS Mice were exposed to 1.5% isoflurane for 4 h at postnatal day 7. The mammalian target of rapamycin inhibitor, rapamycin, or the promyelination drug, clemastine, were administered on days 21 to 35. Mice underwent Y-maze and novel object position recognition tests (n = 12 per group) on days 56 to 62 or were euthanized for either immunohistochemistry (n = 8 per group) or Western blotting (n = 8 per group) at day 35 or were euthanized for electron microscopy at day 63. RESULTS Isoflurane exposure increased the percentage of phospho-S6-positive oligodendrocytes in fimbria of hippocampus from 22 ± 7% to 51 ± 6% (P < 0.0001). In Y-maze testing, isoflurane-exposed mice did not discriminate normally between old and novel arms, spending equal time in both (50 ± 5% old:50 ± 5% novel; P = 0.999), indicating impaired spatial learning. Treatment with clemastine restored discrimination, as evidenced by increased time spent in the novel arm (43 ± 6% old:57 ± 6% novel; P < 0.001), and rapamycin had a similar effect (44 ± 8% old:56 ± 8% novel; P < 0.001). Electron microscopy shows a reduction in myelin thickness as measured by an increase in g-ratio from 0.76 ± 0.06 for controls to 0.79 ± 0.06 for the isoflurane group (P < 0.001). Isoflurane exposure followed by rapamycin treatment resulted in a g-ratio (0.75 ± 0.05) that did not differ significantly from the control value (P = 0.426). Immunohistochemistry and Western blotting show that isoflurane acts on oligodendrocyte precursor cells to inhibit both proliferation and differentiation. DNA methylation and expression of a DNA methyl transferase 1 are reduced in oligodendrocyte precursor cells after isoflurane treatment. Effects of isoflurane on oligodendrocyte precursor cells were abolished by treatment with rapamycin. CONCLUSIONS Early postnatal exposure to isoflurane in mice causes lasting disruptions of oligodendrocyte development in the hippocampus via actions on the mammalian target of rapamycin pathway.
Collapse
|
48
|
Zhang L, Zhuang X, Chen Y, Xia H. Intravenous transplantation of olfactory bulb ensheathing cells for a spinal cord hemisection injury rat model. Cell Transplant 2019; 28:1585-1602. [PMID: 31665910 PMCID: PMC6923555 DOI: 10.1177/0963689719883842] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Cellular transplantation strategies utilizing intraspinal or intrathecal olfactory
ensheathing cells (OECs) have been reported as beneficial for spinal cord injury (SCI).
However, there are many disadvantages of these methods, including additional trauma to the
spinal cord parenchyma and technical challenges. Therefore, we investigated the
feasibility and potential benefits of intravenous transplantation of OECs in a rat
hemisection SCI model. OECs derived from olfactory bulb tissue were labeled with quantum
dots (QDs), and their biodistribution after intravenous transplantation was tracked using
a fluorescence imaging system. Accumulation of the transplanted OECs was observed in the
injured spinal cord within 10 min, peaked at seven days after cell transplantation, and
decreased gradually thereafter. This time window corresponded to the blood–spinal cord
barrier (BSCB) opening time, which was quantitated with the Evans blue leakage assay.
Using immunohistochemistry, we examined neuronal growth (GAP-43), remyelination (MBP), and
microglia (Iba-1) reactions at the lesion site. Motor function recovery was also measured
using a classic open field test (Basso, Beattie and Bresnahan score). Compared with the
group injected only with QDs, the rats that received OEC transplantation exhibited a
prominent reduction in inflammatory responses, increased neurogenesis and remyelination,
and significant improvement in motor function. We suggest that intravenous injection could
also be an effective method for delivering OECs and improving functional outcomes after
SCI. Moreover, the time course of BSCB disruption provides a clinically relevant
therapeutic window for cell-based intervention.
Collapse
Affiliation(s)
- Lijian Zhang
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.,Ningxia Human Stem Cell Research Institute, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.,Surgery Laboratory, General Hospital of Ningxia Medical University, Yinchuan, China.,Both the authors are co-authors and contributed equally to this article
| | - Xiaoqing Zhuang
- Department of Nuclear Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.,Both the authors are co-authors and contributed equally to this article
| | - Yao Chen
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.,Ningxia Human Stem Cell Research Institute, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Hechun Xia
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.,Ningxia Human Stem Cell Research Institute, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
49
|
What Do Microglia Really Do in Healthy Adult Brain? Cells 2019; 8:cells8101293. [PMID: 31652490 PMCID: PMC6829860 DOI: 10.3390/cells8101293] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/17/2019] [Accepted: 10/18/2019] [Indexed: 12/27/2022] Open
Abstract
Microglia originate from yolk sac-primitive macrophages and auto-proliferate into adulthood without replacement by bone marrow-derived circulating cells. In inflammation, stroke, aging, or infection, microglia have been shown to contribute to brain pathology in both deleterious and beneficial ways, which have been studied extensively. However, less is known about their role in the healthy adult brain. Astrocytes and oligodendrocytes are widely accepted to strongly contribute to the maintenance of brain homeostasis and to modulate neuronal function. On the other hand, contribution of microglia to cognition and behavior is only beginning to be understood. The ability to probe their function has become possible using microglial depletion assays and conditional mutants. Studies have shown that the absence of microglia results in cognitive and learning deficits in rodents during development, but this effect is less pronounced in adults. However, evidence suggests that microglia play a role in cognition and learning in adulthood and, at a cellular level, may modulate adult neurogenesis. This review presents the case for repositioning microglia as key contributors to the maintenance of homeostasis and cognitive processes in the healthy adult brain, in addition to their classical role as sentinels coordinating the neuroinflammatory response to tissue damage and disease.
Collapse
|
50
|
Fjell AM, Sneve MH, Sederevicius D, Sørensen Ø, Krogsrud SK, Mowinckel AM, Walhovd KB. Volumetric and microstructural regional changes of the hippocampus underlying development of recall performance after extended retention intervals. Dev Cogn Neurosci 2019; 40:100723. [PMID: 31678691 PMCID: PMC6974909 DOI: 10.1016/j.dcn.2019.100723] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 10/10/2019] [Accepted: 10/19/2019] [Indexed: 01/12/2023] Open
Abstract
Unique developmental effects on recall over days rather than minutes. Development of visual recall explainable by visuo-constructive ability. Development of verbal recall not explained by verbal ability. Modest relationships between recall performance and hippocampus structure.
Performance on recall tests improves through childhood and adolescence, in part due to structural maturation of the medial temporal cortex. Although partly different processes support successful recall over shorter vs. longer intervals, recall is usually tested after less than an hour. The aim of the present study was to test whether there are unique developmental changes in recall performance using extended retention intervals, and whether these are related to structural maturation of sub-regions of the hippocampus. 650 children and adolescents from 4.1 to 24.8 years were assessed in total 962 times (mean interval ≈ 1.8 years). The California Verbal Learning Test (CVLT) and the Rey Complex Figure Test (CFT) were used. Recall was tested 30 min and ≈ 10 days after encoding. We found unique developmental effects on recall in the extended retention interval condition independently of 30 min recall performance. For CVLT, major improvements happened between 10 and 15 years. For CFT, improvement was linear and was accounted for by visuo-constructive abilities. The relationships did not show anterior-posterior hippocampal axis differences. In conclusion, performance on recall tests using extended retention intervals shows unique development, likely due to changes in encoding depth or efficacy, or improvements of long-term consolidation processes.
Collapse
Affiliation(s)
- Anders M Fjell
- Center for Lifespan Changes in Brain and Cognition, University of Oslo, Norway; Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway.
| | - Markus H Sneve
- Center for Lifespan Changes in Brain and Cognition, University of Oslo, Norway
| | | | - Øystein Sørensen
- Center for Lifespan Changes in Brain and Cognition, University of Oslo, Norway
| | - Stine K Krogsrud
- Center for Lifespan Changes in Brain and Cognition, University of Oslo, Norway
| | | | - Kristine B Walhovd
- Center for Lifespan Changes in Brain and Cognition, University of Oslo, Norway; Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|