1
|
Singh AK, Joshi I, Reddy NMN, Purushotham SS, Eswaramoorthy M, Vasudevan M, Banerjee S, Clement JP, Kundu TK. Epigenetic modulation rescues neurodevelopmental deficits in Syngap1 +/- mice. Aging Cell 2025:e14408. [PMID: 39878322 DOI: 10.1111/acel.14408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/05/2024] [Accepted: 10/22/2024] [Indexed: 01/31/2025] Open
Abstract
SYNGAP1 is a Ras GTPase-activating protein that plays a crucial role during brain development and in synaptic plasticity. Sporadic heterozygous mutations in SYNGAP1 affect social and emotional behaviour observed in intellectual disability (ID) and autism spectrum disorder (ASD). Although neurophysiological deficits have been extensively studied, the epigenetic landscape of SYNGAP1 mutation-mediated intellectual disability is unexplored. Here, we have found that the p300/CBP specific acetylation marks of histones are significantly repressed in the hippocampus of adolescent Syngap1+/- mice. Additionally, we observed decreased dendritic branching of newly born DCX+ neurons in these mice, suggesting altered adult hippocampal neurogenesis. To establish the causal relationship of Syngap1+/- phenotype and the altered histone acetylation signature we have treated 2-4 months old Syngap1+/- mice with glucose-derived carbon nanosphere (CSP) conjugated potent small molecule activator (TTK21) of p300/CBP lysine acetyltransferase (CSP-TTK21). The enhancement of the p300/CBP specific acetylation marks of histones by CSP-TTK21 restored synaptic functions, increased dendritic branching of DCX+ neurons, enables the capability to reorganise cortical circuits in response to change in the sensory stimuli, and improves behavioural measures in Syngap1+/- mice that are very closely comparable to wild type littermates. Further, hippocampal RNA-Seq analysis of these mice revealed that the expression of many critical genes such as Adcy1, Ntrk3, Egr1, and Foxj1 which are key regulators of synaptic plasticity and neurogenesis and are well associated with ID/ASD reversed upon CSP-TTK21 treatment. This study could be the first demonstration of the reversal of autistic behaviour and neural wiring upon the modulation of altered epigenetic modification(s).
Collapse
Affiliation(s)
- Akash Kumar Singh
- Molecular Biology and Genetics Unit, Transcription and Disease Laboratory, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Ila Joshi
- Molecular Biology and Genetics Unit, Transcription and Disease Laboratory, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Neeharika M N Reddy
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Sushmitha S Purushotham
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - M Eswaramoorthy
- Chemistry and Physics of Materials Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | | | | | - James P Clement
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Tapas K Kundu
- Molecular Biology and Genetics Unit, Transcription and Disease Laboratory, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| |
Collapse
|
2
|
Zaniewska M, Brygider S, Majcher-Maślanka I, Gawliński D, Głowacka U, Glińska S, Balcerzak Ł. The impact of voluntary wheel-running exercise on hippocampal neurogenesis and behaviours in response to nicotine cessation in rats. Psychopharmacology (Berl) 2024; 241:2585-2607. [PMID: 39463206 PMCID: PMC11569017 DOI: 10.1007/s00213-024-06705-7] [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: 05/29/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024]
Abstract
RATIONALE The literature indicates that nicotine exposure or its discontinuation impair adult hippocampal neurogenesis in rats, though the impact of exercise on this process remains unclear. We have previously shown that disturbances in the number of doublecortin (DCX, a marker of immature neurons)-positive (DCX+) cells in the dentate gyrus (DG) of the hippocampus during nicotine deprivation may contribute to a depression-like state in rats. OBJECTIVES This study aimed to investigate the effect of running on hippocampal neurogenesis, depression-like symptoms, and drug-seeking behaviour during nicotine deprivation. METHODS The rats were subjected to nicotine (0.03 mg/kg/inf) self-administration via an increasing schedule of reinforcement. After 21 sessions, the animals entered a 14-day abstinence phase during which they were housed in either standard home cages without wheels, cages equipped with running wheels, or cages with locked wheels. RESULTS Wheel running increased the number of Ki-67+ and DCX+ cells in the DG of both nicotine-deprived and nicotine-naive rats. Wheel-running exercise evoked an antidepressant effect on abstinence Day 14 but had no effect on nicotine-seeking behaviour on abstinence Day 15 compared to rats with locked-wheel access. CONCLUSIONS In summary, long-term wheel running positively affected the number of immature neurons in the hippocampus, which corresponded with an antidepressant response in nicotine-weaned rats. One possible mechanism underlying the positive effect of running on the affective state during nicotine cessation may be the reduction in deficits in DCX+ cells in the hippocampus.
Collapse
Affiliation(s)
- Magdalena Zaniewska
- Department of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland.
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12 Street, Kraków, 31-343, Poland.
- Affective Cognitive Neuroscience Laboratory, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland.
| | - Sabina Brygider
- Department of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12 Street, Kraków, 31-343, Poland
| | - Iwona Majcher-Maślanka
- Department of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland
| | - Dawid Gawliński
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12 Street, Kraków, 31-343, Poland
| | - Urszula Głowacka
- Department of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, Kraków, 31- 531, Poland
| | - Sława Glińska
- Faculty of Biology and Environmental Protection, Laboratory of Microscopic Imaging and Specialized Biological Techniques, University of Lodz, Banacha 12/16, Lodz, 90-237, Poland
| | - Łucja Balcerzak
- Faculty of Biology and Environmental Protection, Laboratory of Microscopic Imaging and Specialized Biological Techniques, University of Lodz, Banacha 12/16, Lodz, 90-237, Poland
| |
Collapse
|
3
|
Hestehave S, Florea R, Fedorec AJ, Jevic M, Mercy L, Wright A, Morgan OB, Brown LA, Peirson SN, Géranton SM. Differences in multidimensional phenotype of 2 joint pain models link early weight-bearing deficit to late depressive-like behavior in male mice. Pain Rep 2024; 9:e1213. [PMID: 39574483 PMCID: PMC11581759 DOI: 10.1097/pr9.0000000000001213] [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: 05/21/2024] [Revised: 09/11/2024] [Accepted: 09/15/2024] [Indexed: 11/24/2024] Open
Abstract
Introduction Chronic pain is a hallmark of joint diseases. Although these conditions are often accompanied by negative affective symptoms including depression and anxiety, these comorbidities are rarely studied simultaneously in preclinical models where they are poorly characterised. Moreover, how affective symptoms relate to the more obvious sensory and functional symptoms of joint diseases is not well understood. Here, we have addressed these gaps in knowledge. Methods We used 2 preclinical models of joint pain in male mice and an array of behavioural and molecular assays to fully characterise functional deficits, mechanical hypersensitivity, affective symptoms, and nociceptive signaling in joint pain, as well as investigate their relationship. Results Ankle joint inflammation and knee osteoarthritis induced mechanical hypersensitivity that lasted at least 3 months and that was not different between the 2 models on most days. However, the models presented with markedly different weight-bearing deficits, molecular profiles, and affective outcomes. Specifically, only the model of knee osteoarthritis was accompanied by an increase in negative affective behaviors, including early changes in circadian patterns, persistent cognitive impairments, and late development of depressive-like behavior. Importantly, the early weight-bearing deficit strongly correlated with the emotional profiles and the hypersensitivity at 3 months, suggesting that early objective functional measures may be used as predictors of long-term affective symptoms and pain. Conclusion The predictive value of early weight-bearing deficit could prove useful in the clinical setting for adapted therapeutic approaches for the prevention of emotional comorbidities and better pain management for patients with joint pain.
Collapse
Affiliation(s)
- Sara Hestehave
- Department of Cell & Developmental Biology, University College London, London, United Kingdom. Hestehave is now with the Department of Experimental Medicine, University of Copenhagen, Copenhagen N, Denmark
| | - Roxana Florea
- Department of Cell & Developmental Biology, University College London, London, United Kingdom. Hestehave is now with the Department of Experimental Medicine, University of Copenhagen, Copenhagen N, Denmark
| | - Alexander J.H. Fedorec
- Department of Cell & Developmental Biology, University College London, London, United Kingdom. Hestehave is now with the Department of Experimental Medicine, University of Copenhagen, Copenhagen N, Denmark
| | - Maria Jevic
- Department of Cell & Developmental Biology, University College London, London, United Kingdom. Hestehave is now with the Department of Experimental Medicine, University of Copenhagen, Copenhagen N, Denmark
| | - Lucile Mercy
- Department of Cell & Developmental Biology, University College London, London, United Kingdom. Hestehave is now with the Department of Experimental Medicine, University of Copenhagen, Copenhagen N, Denmark
| | - Annia Wright
- Department of Cell & Developmental Biology, University College London, London, United Kingdom. Hestehave is now with the Department of Experimental Medicine, University of Copenhagen, Copenhagen N, Denmark
| | - Oakley B. Morgan
- Department of Cell & Developmental Biology, University College London, London, United Kingdom. Hestehave is now with the Department of Experimental Medicine, University of Copenhagen, Copenhagen N, Denmark
| | | | - Stuart N. Peirson
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, United Kingdom
| | - Sandrine M. Géranton
- Department of Cell & Developmental Biology, University College London, London, United Kingdom. Hestehave is now with the Department of Experimental Medicine, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
4
|
Chauquet S, Willis EF, Grice L, Harley SBR, Powell JE, Wray NR, Nguyen Q, Ruitenberg MJ, Shah S, Vukovic J. Exercise rejuvenates microglia and reverses T cell accumulation in the aged female mouse brain. Aging Cell 2024; 23:e14172. [PMID: 38747044 PMCID: PMC11258432 DOI: 10.1111/acel.14172] [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: 08/16/2023] [Revised: 02/16/2024] [Accepted: 04/01/2024] [Indexed: 07/21/2024] Open
Abstract
Slowing and/or reversing brain ageing may alleviate cognitive impairments. Previous studies have found that exercise may mitigate cognitive decline, but the mechanisms underlying this remain largely unclear. Here we provide unbiased analyses of single-cell RNA sequencing data, showing the impacts of exercise and ageing on specific cell types in the mouse hippocampus. We demonstrate that exercise has a profound and selective effect on aged microglia, reverting their gene expression signature to that of young microglia. Pharmacologic depletion of microglia further demonstrated that these cells are required for the stimulatory effects of exercise on hippocampal neurogenesis but not cognition. Strikingly, allowing 18-month-old mice access to a running wheel did by and large also prevent and/or revert T cell presence in the ageing hippocampus. Taken together, our data highlight the profound impact of exercise in rejuvenating aged microglia, associated pro-neurogenic effects and on peripheral immune cell presence in the ageing female mouse brain.
Collapse
Affiliation(s)
- Solal Chauquet
- Institute for Molecular Bioscience, the University of QueenslandSaint LuciaQueenslandAustralia
| | - Emily F. Willis
- School of Biomedical Sciences, Faculty of MedicineThe University of QueenslandSaint LuciaQueenslandAustralia
| | - Laura Grice
- Institute for Molecular Bioscience, the University of QueenslandSaint LuciaQueenslandAustralia
| | - Samuel B. R. Harley
- Queensland Brain Institute, the University of QueenslandSaint LuciaQueenslandAustralia
| | - Joseph E. Powell
- Institute for Molecular Bioscience, the University of QueenslandSaint LuciaQueenslandAustralia
| | - Naomi R. Wray
- Institute for Molecular Bioscience, the University of QueenslandSaint LuciaQueenslandAustralia
- Department of PsychiatryUniversity of OxfordOxfordUK
- Oxford Big Data Institute, Li Ka Shing Centre for Health Information and DiscoveryUniversity of OxfordOxfordUK
| | - Quan Nguyen
- Institute for Molecular Bioscience, the University of QueenslandSaint LuciaQueenslandAustralia
| | - Marc J. Ruitenberg
- School of Biomedical Sciences, Faculty of MedicineThe University of QueenslandSaint LuciaQueenslandAustralia
| | - Sonia Shah
- Institute for Molecular Bioscience, the University of QueenslandSaint LuciaQueenslandAustralia
| | - Jana Vukovic
- School of Biomedical Sciences, Faculty of MedicineThe University of QueenslandSaint LuciaQueenslandAustralia
- Queensland Brain Institute, the University of QueenslandSaint LuciaQueenslandAustralia
| |
Collapse
|
5
|
Barrett E, Ivey G, Cunningham A, Coffman G, Pemberton T, Lee C, Patra P, Day JB, Lee PHU, Shim JW. Reduced GLP-1R availability in the caudate nucleus with Alzheimer's disease. Front Aging Neurosci 2024; 16:1350239. [PMID: 38915346 PMCID: PMC11194438 DOI: 10.3389/fnagi.2024.1350239] [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: 12/05/2023] [Accepted: 05/15/2024] [Indexed: 06/26/2024] Open
Abstract
The glucagon-like peptide-1 receptor (GLP-1R) agonists reduce glycated hemoglobin in patients with type 2 diabetes. Mounting evidence indicates that the potential of GLP-1R agonists, mimicking a 30 amino acid ligand, GLP-1, extends to the treatment of neurodegenerative conditions, with a particular focus on Alzheimer's disease (AD). However, the mechanism that underlies regulation of GLP-1R availability in the brain with AD remains poorly understood. Here, using whole transcriptome RNA-Seq of the human postmortem caudate nucleus with AD and chronic hydrocephalus (CH) in the elderly, we found that GLP-1R and select mRNAs expressed in glucose dysmetabolism and dyslipidemia were significantly altered. Furthermore, we detected human RNA indicating a deficiency in doublecortin (DCX) levels and the presence of ferroptosis in the caudate nucleus impacted by AD. Using the genome data viewer, we assessed mutability of GLP-1R and 39 other genes by two factors associated with high mutation rates in chromosomes of four species. Surprisingly, we identified that nucleotide sizes of GLP-1R transcript exceptionally differed in all four species of humans, chimpanzees, rats, and mice by up to 6-fold. Taken together, the protein network database analysis suggests that reduced GLP-1R in the aged human brain is associated with glucose dysmetabolism, ferroptosis, and reduced DCX+ neurons, that may contribute to AD.
Collapse
Affiliation(s)
- Emma Barrett
- Department of Biomedical Engineering, Marshall University, Huntington, WV, United States
| | - Gabrielle Ivey
- Department of Biomedical Engineering, Marshall University, Huntington, WV, United States
| | - Adam Cunningham
- Department of Biomedical Engineering, Marshall University, Huntington, WV, United States
| | - Gary Coffman
- Department of Biomedical Engineering, Marshall University, Huntington, WV, United States
| | - Tyera Pemberton
- Department of Biomedical Engineering, Marshall University, Huntington, WV, United States
| | - Chan Lee
- Department of Anesthesia, Indiana University Health Arnett Hospital, Lafayette, IN, United States
| | - Prabir Patra
- Department of Biomedical Engineering, Marshall University, Huntington, WV, United States
| | - James B. Day
- Department of Orthopedic Surgery, Cabell Huntington Hospital and Marshall University School of Medicine, Huntington, WV, United States
| | - Peter H. U. Lee
- Department of Cardiothoracic Surgery, Southcoast Health, Fall River, MA, United States
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Joon W. Shim
- Department of Biomedical Engineering, Marshall University, Huntington, WV, United States
| |
Collapse
|
6
|
Parishar P, Rajagopalan M, Iyengar S. Changes in the dopaminergic circuitry and adult neurogenesis linked to reinforcement learning in corvids. Front Neurosci 2024; 18:1359874. [PMID: 38808028 PMCID: PMC11130420 DOI: 10.3389/fnins.2024.1359874] [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: 02/07/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024] Open
Abstract
The caudolateral nidopallium (NCL, an analog of the prefrontal cortex) is known to be involved in learning, memory, and discrimination in corvids (a songbird), whereas the involvement of other brain regions in these phenomena is not well explored. We used house crows (Corvus splendens) to explore the neural correlates of learning and decision-making by initially training them on a shape discrimination task followed by immunohistochemistry to study the immediate early gene expression (Arc), a dopaminoceptive neuronal marker (DARPP-32, Dopamine- and cAMP-regulated phosphoprotein, Mr 32 kDa) to understand the involvement of the reward pathway and an immature neuronal marker (DCX, doublecortin) to detect learning-induced changes in adult neurogenesis. We performed neuronal counts and neuronal tracing, followed by morphometric analyses. Our present results have demonstrated that besides NCL, other parts of the caudal nidopallium (NC), avian basal ganglia, and intriguingly, vocal control regions in house crows are involved in visual discrimination. We have also found that training on the visual discrimination task can be correlated with neurite pruning in mature dopaminoceptive neurons and immature DCX-positive neurons in the NC of house crows. Furthermore, there is an increase in the incorporation of new neurons throughout NC and the medial striatum which can also be linked to learning. For the first time, our results demonstrate that a combination of structural changes in mature and immature neurons and adult neurogenesis are linked to learning in corvids.
Collapse
|
7
|
Davila-Valencia I, Saad M, Olthoff G, Faulkner M, Charara M, Farnum A, Dysko RC, Zhang Z. Sex specific effects of buprenorphine on adult hippocampal neurogenesis and behavioral outcomes during the acute phase after pediatric traumatic brain injury in mice. Neuropharmacology 2024; 245:109829. [PMID: 38159797 DOI: 10.1016/j.neuropharm.2023.109829] [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: 10/05/2023] [Revised: 12/15/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
Traumatic brain injury (TBI) in children often causes cognitive and mental dysfunctions, as well as acute and chronic pain. Adult hippocampal neurogenesis plays a key role in cognition, depression, and pain. Adult hippocampal neurogenesis can be modulated by genetic and environmental factors, such as TBI and opioids. Buprenorphine (BPN), a semisynthetic opioid, is commonly used for pain management in children, however, the effects of BPN on adult hippocampal neurogenesis after pediatric TBI are still unclear. This study investigated the sex-specific effects of BPN on adult hippocampal neurogenesis during acute phase after pediatric TBI. Male and female littermates were randomized on postnatal day 20-21(P20-21) into Sham, TBI+saline and TBI+BPN groups. BPN was administered intraperitoneally to the TBI+BPN mice at 30 min after injury, and then every 6-12 h (h) for 2 days (d). Bromodeoxyuridine (BrdU) was administered intraperitoneally to all groups at 2, 4, 6, and 8-h post-injury. All outcomes were evaluated at 3-d post-BrdU administration. We found that TBI induced significant cognitive impairment, depression, and reduced adult hippocampal neurogenesis in both male and female mice, with more prominent effects in females. BPN significantly improved adult hippocampal neurogenesis and depression in males, but not in females. We further demonstrated that differential expressions of opioid receptors, transcription factors and neuroinflammatory markers at the neurogenic niche might be responsible for the differential effects of BPN in males and females. In conclusion, this study elucidates the effects of BPN on adult hippocampal neurogenesis and behavioral outcomes at the acute phase after pediatric TBI.
Collapse
Affiliation(s)
- Ivan Davila-Valencia
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Mark Saad
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Grace Olthoff
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Megan Faulkner
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Maysoun Charara
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Abigail Farnum
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Robert C Dysko
- Unit for Laboratory Animal Medicine, University of Michigan-Ann Arbor, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA.
| | - Zhi Zhang
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| |
Collapse
|
8
|
Willis EF, Kim SJ, Chen W, Nyuydzefe M, MacDonald KPA, Zanin-Zhorov A, Ruitenberg MJ, Vukovic J. ROCK2 regulates microglia proliferation and neuronal survival after traumatic brain injury. Brain Behav Immun 2024; 117:181-194. [PMID: 38211634 DOI: 10.1016/j.bbi.2024.01.004] [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: 07/14/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/13/2024] Open
Abstract
Traumatic brain injury (TBI) results in prolonged and non-resolving activation of microglia. Forced turnover of these cells during the acute phase of TBI aids recovery, but the cell-intrinsic pathways that underpin the pro-repair phenotype of these repopulating microglia remain unclear. Here, we show that selective targeting of ROCK2 with the small molecule inhibitor KD025 impairs the proliferative response of microglia after TBI as well as during genetically induced turnover of microglia. KD025 treatment abolished the substantial neuroprotective and cognitive benefits conferred by repopulating microglia, preventing these cells from replenishing the depleted niche during the early critical time window post-injury. Delaying KD025 treatment to the subacute phase of TBI allowed microglial repopulation to occur, but this did not enhance the benefits conferred by repopulating microglia. Taken together, our data indicate that ROCK2 mediates neuronal survival and microglial population dynamics after TBI, including the emergence of repopulating microglia with a pro-repair phenotype.
Collapse
Affiliation(s)
- Emily F Willis
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Australia
| | - Seung Jae Kim
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Australia
| | - Wei Chen
- Graviton Bioscience Corporation, Gravition Bioscience B.V., Amsterdam, Netherlands
| | - Melanie Nyuydzefe
- Graviton Bioscience Corporation, Gravition Bioscience B.V., Amsterdam, Netherlands
| | | | | | - Marc J Ruitenberg
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Australia
| | - Jana Vukovic
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Australia; Queensland Brain Institute, The University of Queensland, Australia.
| |
Collapse
|
9
|
Shaikh SN, Willis EF, Dierich M, Xu Y, Stuart SJS, Gobe GC, Bashaw AA, Rawashdeh O, Kim SJ, Vukovic J. CSF-1R inhibitor PLX3397 attenuates peripheral and brain chronic GVHD and improves functional outcomes in mice. J Neuroinflammation 2023; 20:300. [PMID: 38102698 PMCID: PMC10725001 DOI: 10.1186/s12974-023-02984-7] [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: 08/16/2023] [Accepted: 12/03/2023] [Indexed: 12/17/2023] Open
Abstract
Graft-versus-host disease (GVHD) is a serious complication of otherwise curative allogeneic haematopoietic stem cell transplants. Chronic GVHD induces pathological changes in peripheral organs as well as the brain and is a frequent cause of late morbidity and death after bone-marrow transplantation. In the periphery, bone-marrow-derived macrophages are key drivers of pathology, but recent evidence suggests that these cells also infiltrate into cGVHD-affected brains. Microglia are also persistently activated in the cGVHD-affected brain. To understand the involvement of these myeloid cell populations in the development and/or progression of cGVHD pathology, we here utilized the blood-brain-barrier permeable colony stimulating factor-1 receptor (CSF-1R) inhibitor PLX3397 (pexidartinib) at varying doses to pharmacologically deplete both cell types. We demonstrate that PLX3397 treatment during the development of cGVHD (i.e., 30 days post-transplant) improves disease symptoms, reducing both the clinical scores and histopathology of multiple cGVHD target organs, including the sequestration of T cells in cGVHD-affected skin tissue. Cognitive impairments associated with cGVHD and neuroinflammation were also attenuated by PLX3397 treatment. PLX3397 treatment prior to the onset of cGVHD (i.e., immediately post-transplant) did not change in clinical scores or histopathology. Overall, our data demonstrate significant benefits of using PLX3397 for the treatment of cGVHD and associated organ pathologies in both the periphery and brain, highlighting the therapeutic potential of pexidartinib for this condition.
Collapse
Affiliation(s)
- Samreen N Shaikh
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| | - Emily F Willis
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| | - Max Dierich
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| | - Yi Xu
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| | - Samuel J S Stuart
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| | - Glenda C Gobe
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| | - Abate A Bashaw
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| | - Oliver Rawashdeh
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| | - Seung Jae Kim
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| | - Jana Vukovic
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia.
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
10
|
Kamte YS, Chandwani MN, London NM, Potosnak CE, Leak RK, O'Donnell LA. Perturbations in neural stem cell function during a neurotropic viral infection in juvenile mice. J Neurochem 2023; 166:809-829. [PMID: 37530081 DOI: 10.1111/jnc.15914] [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: 03/17/2023] [Revised: 06/25/2023] [Accepted: 06/28/2023] [Indexed: 08/03/2023]
Abstract
Viral infections of the central nervous system (CNS) often cause worse neurological outcomes in younger hosts. Throughout childhood, the brain undergoes extensive development and refinement to produce functional neural networks. Network function is maintained partly with the help of neural stem cells (NSCs) that replace neuronal and glia subtypes in the two neurogenic niches of the brain (the hippocampus and subventricular zone). Accumulating evidence suggests that viruses disrupt NSC function in adulthood and infancy, but the in vivo impact of childhood infections on acute and long-term NSC function is unknown. Using a juvenile mouse model of measles virus (MeV) infection, where only mature neurons in the brain are infected, we defined the effects of the antiviral immune response on NSCs from juvenile to adult stages of life. We found that (a) virus persists in the brains of survivors despite an anti-viral immune response; (b) NSC numbers decrease dramatically during early infection, but ultimately stabilize in adult survivors; (c) infection is associated with mild apoptosis throughout the juvenile brain, but NSC proliferation is unchanged; (d) the loss of NSC numbers is dependent upon the stage of NSC differentiation; and (e) immature neurons increase early during infection, concurrent with depletion of NSC pools. Collectively, we show that NSCs are exquisitely sensitive to the inflammatory microenvironment created during neuron-restricted MeV infection in juveniles, responding with an early loss of NSCs but increased neurogenesis. These studies provide insight into potential cellular mechanisms associated with long-term neurological deficits in survivors of childhood CNS infections.
Collapse
Affiliation(s)
- Yashika S Kamte
- School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Manisha N Chandwani
- School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Natalie M London
- School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Chloe E Potosnak
- School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Rehana K Leak
- School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Lauren A O'Donnell
- School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
11
|
Leiter O, Brici D, Fletcher SJ, Yong XLH, Widagdo J, Matigian N, Schroer AB, Bieri G, Blackmore DG, Bartlett PF, Anggono V, Villeda SA, Walker TL. Platelet-derived exerkine CXCL4/platelet factor 4 rejuvenates hippocampal neurogenesis and restores cognitive function in aged mice. Nat Commun 2023; 14:4375. [PMID: 37587147 PMCID: PMC10432533 DOI: 10.1038/s41467-023-39873-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 06/20/2023] [Indexed: 08/18/2023] Open
Abstract
The beneficial effects of physical activity on brain ageing are well recognised, with exerkines, factors that are secreted into the circulation in response to exercise, emerging as likely mediators of this response. However, the source and identity of these exerkines remain unclear. Here we provide evidence that an anti-geronic exerkine is secreted by platelets. We show that platelets are activated by exercise and are required for the exercise-induced increase in hippocampal precursor cell proliferation in aged mice. We also demonstrate that increasing the systemic levels of the platelet-derived exerkine CXCL4/platelet factor 4 (PF4) ameliorates age-related regenerative and cognitive impairments in a hippocampal neurogenesis-dependent manner. Together these findings highlight the role of platelets in mediating the rejuvenating effects of exercise during physiological brain ageing.
Collapse
Affiliation(s)
- Odette Leiter
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - David Brici
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Stephen J Fletcher
- Centre for Horticultural Science, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Xuan Ling Hilary Yong
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Jocelyn Widagdo
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Nicholas Matigian
- Queensland Cyber Infrastructure Foundation Ltd, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Adam B Schroer
- Department of Anatomy, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Gregor Bieri
- Department of Anatomy, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Daniel G Blackmore
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Perry F Bartlett
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Victor Anggono
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Saul A Villeda
- Department of Anatomy, University of California San Francisco, San Francisco, CA, 94143, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, 94143, USA
- Bakar Aging Research Institute, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Tara L Walker
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
12
|
Willis EF, Gillespie ER, Guse K, Zuercher AW, Käsermann F, Ruitenberg MJ, Vukovic J. Intravenous immunoglobulin (IVIG) promotes brain repair and improves cognitive outcomes after traumatic brain injury in a FcγRIIB receptor-dependent manner. Brain Behav Immun 2023; 109:37-50. [PMID: 36581304 DOI: 10.1016/j.bbi.2022.12.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 12/13/2022] [Accepted: 12/23/2022] [Indexed: 12/27/2022] Open
Abstract
Intravenous immunoglobulin (IVIG) is a promising immune-modulatory therapy for limiting harmful inflammation and associated secondary tissue loss in neurotrauma. Here, we show that IVIG therapy attenuates spatial learning and memory deficits following a controlled cortical impact mouse model of traumatic brain injury (TBI). These improvements in cognitive outcomes were associated with increased neuronal survival, an overall reduction in brain tissue loss, and a greater preservation of neural connectivity. Furthermore, we demonstrate that the presence of the main inhibitory FcγRIIB receptor is required for the beneficial effects of IVIG treatment in TBI, with our results simultaneously highlighting the role of this receptor in reducing secondary damage arising from brain injury.
Collapse
Affiliation(s)
- Emily F Willis
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Ellen R Gillespie
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Kirsten Guse
- CSL Behring, Research, CSL Biologics Research Center, Bern, Switzerland
| | - Adrian W Zuercher
- CSL Behring, Research, CSL Biologics Research Center, Bern, Switzerland
| | - Fabian Käsermann
- CSL Behring, Research, CSL Biologics Research Center, Bern, Switzerland
| | - Marc J Ruitenberg
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Jana Vukovic
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia; Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
13
|
Blackmore DG, Waters MJ. The multiple roles of GH in neural ageing and injury. Front Neurosci 2023; 17:1082449. [PMID: 36960169 PMCID: PMC10027725 DOI: 10.3389/fnins.2023.1082449] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 02/17/2023] [Indexed: 03/09/2023] Open
Abstract
Advanced age is typically associated with a decrease in cognitive function including impairment in the formation and retention of new memories. The hippocampus is critical for learning and memory, especially spatial learning, and is particularly affected by ageing. With advanced age, multiple neural components can be detrimentally affected including a reduction in the number of neural stem and precursor cells, a decrease in the formation of adult born neurons (neurogenesis), and deficits in neural circuitry, all of which ultimately contribute to impaired cognitive function. Importantly, physical exercise has been shown to ameliorate many of these impairments and is able to improve learning and memory. Relevantly, growth hormone (GH) is an important protein hormone that decreases with ageing and increases following physical exercise. Originally described due to its role in longitudinal growth, GH has now been identified to play several additional key roles, especially in relation to the brain. Indeed, the regular decrease in GH levels following puberty is one of the most well documented components of neuroendocrine ageing. Growth hormone deficiency (GHD) has been described to have adverse effects on brain function, which can be ameliorated via GH replacement therapy. Physical exercise has been shown to increase circulating GH levels. Furthermore, we recently demonstrated the increase in exercise-mediated GH is critical for improved cognitive function in the aged mouse. Here we examine the multiple roles that GH plays, particularly in the aged brain and following trauma, irradiation and stroke, and how increasing GH levels can ameliorate deficits in cognition.
Collapse
Affiliation(s)
- Daniel G. Blackmore
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Michael J. Waters
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- *Correspondence: Michael J. Waters,
| |
Collapse
|
14
|
Mateo E, Tonino RPB, Canto A, Monroy Noyola A, Miranda M, Soria JM, Garcia Esparza MA. The Neurotoxic Effect of Ochratoxin-A on the Hippocampal Neurogenic Niche of Adult Mouse Brain. Toxins (Basel) 2022; 14:624. [PMID: 36136561 PMCID: PMC9501519 DOI: 10.3390/toxins14090624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/24/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Ochratoxin A (OTA) is a common secondary metabolite of Aspergillus ochraceus, A. carbonarius, and Penicillium verrucosum. This mycotoxin is largely present as a contaminant in several cereal crops and human foodstuffs, including grapes, corn, nuts, and figs, among others. Preclinical studies have reported the involvement of OTA in metabolic, physiologic, and immunologic disturbances as well as in carcinogenesis. More recently, it has also been suggested that OTA may impair hippocampal neurogenesis in vivo and that this might be associated with learning and memory deficits. Furthermore, aside from its widely proven toxicity in tissues other than the brain, there is reason to believe that OTA contributes to neurodegenerative disorders. Thus, in this present in vivo study, we investigated this possibility by intraperitoneally (i.p.) administering 3.5 mg OTA/kg body weight to adult male mice to assess whether chronic exposure to this mycotoxin negatively affects cell viability in the dentate gyrus of the hippocampus. Immunohistochemistry assays showed that doses of 3.5 mg/kg caused a significant and dose-dependent reduction in repetitive cell division and branching (from 12% to 62%). Moreover, the number of countable astrocytes (p < 0.001), young neurons (p < 0.001), and mature neurons (p < 0.001) negatively correlated with the number of i.p. OTA injections administered (one, two, three, or six repeated doses). Our results show that OTA induced adverse effects in the hippocampus cells of adult mice brain tissue when administered in cumulative doses.
Collapse
Affiliation(s)
- Eva Mateo
- Department of Microbiology and Ecology, School of Medicine and Dentistry, University of Valencia, 46001 Valencia, Spain
| | | | - Antolin Canto
- Department of Biomedical Sciences, Cardenal Herrera University-CEU Universities, 46001 Valencia, Spain
| | - Antonio Monroy Noyola
- Neuroprotection Laboratory, Faculty of Pharmacy, Autonomous University of the State of Morelos, Cuernavaca, Morelos 98100, Mexico
| | - Maria Miranda
- Department of Biomedical Sciences, Cardenal Herrera University-CEU Universities, 46001 Valencia, Spain
| | - Jose Miguel Soria
- Department of Biomedical Sciences, Cardenal Herrera University-CEU Universities, 46001 Valencia, Spain
| | | |
Collapse
|
15
|
Protocol for three alternative paradigms to test spatial learning and memory in mice. STAR Protoc 2022; 3:101500. [PMID: 35776637 PMCID: PMC9249832 DOI: 10.1016/j.xpro.2022.101500] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/04/2022] [Accepted: 06/07/2022] [Indexed: 11/23/2022] Open
|
16
|
5 Hz of repetitive transcranial magnetic stimulation improves cognition and induces modifications in hippocampal neurogenesis in adult female Swiss Webster mice. Brain Res Bull 2022; 186:91-105. [PMID: 35688304 DOI: 10.1016/j.brainresbull.2022.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/23/2022] [Accepted: 06/03/2022] [Indexed: 11/22/2022]
Abstract
Adult hippocampal neurogenesis is regulated by several stimuli to promote the creation of a reserve that may facilitate coping with environmental challenges. In this regard, repetitive transcranial magnetic stimulation (rTMS), a neuromodulation therapy, came to our attention because in clinical studies it reverts behavioral and cognitive alterations related to changes in brain plasticity. Some preclinical studies emphasize the need to understand the underlying mechanism of rTMS to induce behavioral modifications. In this study, we investigated the effects of rTMS on cognition, neurogenic-associated modifications, and neuronal activation in the hippocampus of female Swiss Webster mice. We applied 5 Hz of rTMS twice a day for 14 days. Three days later, mice were exposed to the behavioral battery. Then, brains were collected and immunostained for Ki67-positive cells, doublecortin-positive (DCX+)-cells, calbindin, c-Fos and FosB/Delta-FosB in the dentate gyrus. Also, we analyzed mossy fibers and CA3 with calbindin immunostaining. Mice exposed to rTMS exhibited cognitive improvement, an increased number of proliferative cells, DCX cells, DCX cells with complex dendrite morphology, c-Fos and immunoreactivity of FosB/Delta-FosB in the granular cell layer. The volume of the granular cell layer, mossy fibers and CA3 in rTMS mice also increased. Interestingly, cognitive improvement correlated with DCX cells with complex dendrite morphology. Also, those DCX cells and calbindin immunoreactivity correlated with c-Fos in the granular cell layer. Our results suggest that 5 Hz of rTMS applied twice a day modify cell proliferation, doublecortin cells, mossy fibers and enhance cognitive behavior in healthy female Swiss Webster mice.
Collapse
|
17
|
Wang Y, Wen W, Li H, Xu H, Xu M, Ma M, Luo J. Deficiency of mesencephalic astrocyte-derived neurotrophic factor affects neurogenesis in mouse brain. Brain Res Bull 2022; 183:49-56. [PMID: 35227768 PMCID: PMC10014018 DOI: 10.1016/j.brainresbull.2022.02.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/18/2022] [Accepted: 02/24/2022] [Indexed: 12/31/2022]
Abstract
The mechanisms underlying the regulation of neurogenesis in the adult brain remain unclear. Mesencephalic astrocyte-derived neurotrophic factor (MANF) is a neurotrophic factor that has been implicated in various neuropathological processes and endoplasmic reticulum stress. However, the role of MANF in neurogenesis has not been investigated. Using a central nervous system (CNS)-specific Manf knock-out mouse model, we examined the role of MANF in mouse neurogenesis. We demonstrated that MANF deficiency increased BrdU labeling and Ki-67 positive cells in the subgranular zone and subventricular zone. MANF knock-out-induced upregulation of proliferative activity was accompanied by a decrease of cell cycle inhibitors (p15 and p27), an increase of G2/M marker (phospho-histone H3), as well as an increase of neural progenitor markers (Sox2 and NeuroD1) in the brain. In vitro studies using N2A neuroblastoma cells showed that the gain-of-function of MANF inhibited cell cycle progression, whereas the loss-of-function of MANF promoted cell cycle progression. Collectively, our findings indicate MANF deficiency affects cell proliferation and suggest a role of MANF in the neurogenesis of the adult brain.
Collapse
Affiliation(s)
- Yongchao Wang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37372, USA
| | - Wen Wen
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Hui Li
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Hong Xu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Mei Xu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Murong Ma
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Jia Luo
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa City VA Health Care System, Iowa City, IA 52246, USA.
| |
Collapse
|
18
|
Xie WS, Shehzadi K, Ma HL, Liang JH. A Potential Strategy for Treatment of Neurodegenerative Disorders by Regulation of Adult Hippocampal Neurogenesis in Human Brain. Curr Med Chem 2022; 29:5315-5347. [DOI: 10.2174/0929867329666220509114232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/13/2022] [Accepted: 03/17/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
Adult hippocampal neurogenesis is a multistage mechanism that continues throughout the lifespan of human and non-human mammals. These adult-born neurons in the central nervous system (CNS) play a significant role in various hippocampus-dependent processes, including learning, mood regulation, pattern recognition, etc. Reduction of adult hippocampal neurogenesis, caused by multiple factors such as neurological disorders and aging, would impair neuronal proliferation and differentiation and result in memory loss. Accumulating studies have indicated that functional neuron impairment could be restored by promoting adult hippocampal neurogenesis. In this review, we summarized the small molecules that could efficiently promote the process of adult neurogenesis, particularly the agents that have the capacity of crossing the blood-brain barrier (BBB), and showed in vivo efficacy in mammalian brains. This may pave the way for the rational design of drugs to treat humnan neurodegenerative disorders in the future.
Collapse
Affiliation(s)
- Wei-Song Xie
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
| | - Kiran Shehzadi
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
| | - Hong-Le Ma
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
| | - Jian-Hua Liang
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
- Yangtze Delta Region Academy of Beijing Institute of Technology, Jiaxing 314019, China
| |
Collapse
|
19
|
Attenuation of isoprenaline-induced myocardial infarction by Rheum turkestanicum. Biomed Pharmacother 2022; 148:112775. [PMID: 35240528 DOI: 10.1016/j.biopha.2022.112775] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/13/2022] [Accepted: 02/27/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Oxidative stress plays a major role in the pathogenesis of myocardial infarction. This study evaluated the cardioprotective effects of the hydroalcoholic extract of Rheum turkestanicum on isoprenaline-induced myocardial infarction (MI) in Wistar rats. METHODS In this study, we used liquid chromatography-mass spectrometry to determine the active compounds present in the extract. Thirty rats were divided to 5 groups (6 rats in each group). The extract was administered orally at the doses of 100 and 300 mg/kg body weight and then a subcutaneous injection of isoprenaline (85 mg/kg) was administered on the 8th and 9th days. Serum levels of lactate dehydrogenase (LDH), creatine kinase-MB (CK-MB), and creatinine kinase (CPK) were measured using standard commercial kits. Serum activities of superoxide dismutase, catalase, and cardiac levels of thiol and lipid peroxidation were also determined. Hematoxylin and eosin were used for histopathological staining. RESULTS Phytochemical analysis revealed the presence of 24 compounds in the hydro-ethanolic extract of R. turkestanicum. Isoprenaline increased malondialdehyde (4.002 ± 0178, P < 0.001) while decreased thiol content (101.7 ± 6.186, P < 0.001). Moreover, reduced activities of superoxide dismutase (139 ± 10.88, P < 0.001) and catalase (2.812 ± 0.215, P < 0.001), and elevated levels of LDH (1245 ± 62.28, P < 0.001), CPK (898 ± 23.06, P < 0.001) and CK-MB (697 ± 50.22, P < 0.001) were observed. Pretreatment with the R. turkestanicum extract significantly reduced cardiac markers and increased thiol content as well as the activity of antioxidant enzymes. The extract attenuated the histopathological changes induced by isoprenaline. CONCLUSION According to the obtained results, R. turkestanicum may be an appropriate candidate to reduce isoprenaline-induced MI through modulation of oxidative stress. Administration of the extract attenuated cardiac enzymes following isoprenaline administration. The cardioprotective action of the extract can be attributed to the bioactive antioxidant ingredients of R. turkestanicum. To identify the precise mechanisms, further investigations are required.
Collapse
|
20
|
Adams RC, Carter-Cusack D, Shaikh SN, Llanes GT, Johnston RL, Quaife-Ryan G, Boyle G, Koufariotis LT, Möller A, Blazar BR, Vukovic J, MacDonald KPA. Donor bone marrow-derived macrophage MHC II drives neuroinflammation and altered behavior during chronic GVHD in mice. Blood 2022; 139:1389-1408. [PMID: 34570880 PMCID: PMC8900272 DOI: 10.1182/blood.2021011671] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 09/03/2021] [Indexed: 11/20/2022] Open
Abstract
Graft-versus-host disease (GVHD) remains the leading cause of nonrelapse mortality after allogeneic stem cell transplantation for hematological malignancies. Manifestations of GVHD in the central nervous system (CNS) present as neurocognitive dysfunction in up to 60% of patients; however, the mechanisms driving chronic GVHD (cGVHD) in the CNS are yet to be elucidated. Our studies of murine cGVHD revealed behavioral deficits associated with broad neuroinflammation and persistent Ifng upregulation. By flow cytometry, we observed a proportional shift in the donor-derived T-cell population in the cGVHD brain from early CD8 dominance to later CD4 sequestration. RNA sequencing of the hippocampus identified perturbations to structural and functional synapse-related gene expression, together with the upregulation of genes associated with interferon-γ responses and antigen presentation. Neuroinflammation in the cortex of mice and humans during acute GVHD was recently shown to be mediated by resident microglia-derived tumor necrosis factor. In contrast, infiltration of proinflammatory major histocompatibility complex (MHC) class II+ donor bone marrow (BM)-derived macrophages (BMDMs) was identified as a distinguishing feature of CNS cGVHD. Donor BMDMs, which composed up to 50% of the CNS myeloid population, exhibited a transcriptional signature distinct from resident microglia. Recipients of MHC class II knockout BM grafts exhibited attenuated neuroinflammation and behavior comparable to controls, suggestive of a critical role of donor BMDM MHC class II expression in CNS cGVHD. Our identification of disease mediators distinct from those in the acute phase indicates the necessity to pursue alternative therapeutic targets for late-stage neurological manifestations.
Collapse
Affiliation(s)
- Rachael C Adams
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Faculty of Medicine, School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Dylan Carter-Cusack
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Samreen N Shaikh
- Faculty of Medicine, School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Genesis T Llanes
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Rebecca L Johnston
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Gregory Quaife-Ryan
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Glen Boyle
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Lambros T Koufariotis
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Andreas Möller
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Bruce R Blazar
- Masonic Cancer Center and
- Division of Blood & Marrow Transplant & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, MN; and
| | - Jana Vukovic
- Faculty of Medicine, School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
| | - Kelli P A MacDonald
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
21
|
Leiter O, Zhuo Z, Rust R, Wasielewska JM, Grönnert L, Kowal S, Overall RW, Adusumilli VS, Blackmore DG, Southon A, Ganio K, McDevitt CA, Rund N, Brici D, Mudiyan IA, Sykes AM, Rünker AE, Zocher S, Ayton S, Bush AI, Bartlett PF, Hou ST, Kempermann G, Walker TL. Selenium mediates exercise-induced adult neurogenesis and reverses learning deficits induced by hippocampal injury and aging. Cell Metab 2022; 34:408-423.e8. [PMID: 35120590 DOI: 10.1016/j.cmet.2022.01.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/01/2021] [Accepted: 01/11/2022] [Indexed: 12/24/2022]
Abstract
Although the neurogenesis-enhancing effects of exercise have been extensively studied, the molecular mechanisms underlying this response remain unclear. Here, we propose that this is mediated by the exercise-induced systemic release of the antioxidant selenium transport protein, selenoprotein P (SEPP1). Using knockout mouse models, we confirmed that SEPP1 and its receptor low-density lipoprotein receptor-related protein 8 (LRP8) are required for the exercise-induced increase in adult hippocampal neurogenesis. In vivo selenium infusion increased hippocampal neural precursor cell (NPC) proliferation and adult neurogenesis. Mimicking the effect of exercise through dietary selenium supplementation restored neurogenesis and reversed the cognitive decline associated with aging and hippocampal injury, suggesting potential therapeutic relevance. These results provide a molecular mechanism linking exercise-induced changes in the systemic environment to the activation of quiescent hippocampal NPCs and their subsequent recruitment into the neurogenic trajectory.
Collapse
Affiliation(s)
- Odette Leiter
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia; CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Zhan Zhuo
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia; Brain Research Centre and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Ruslan Rust
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Joanna M Wasielewska
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Lisa Grönnert
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Susann Kowal
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Rupert W Overall
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Vijay S Adusumilli
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Daniel G Blackmore
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Adam Southon
- The Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Katherine Ganio
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Australia
| | - Christopher A McDevitt
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Australia
| | - Nicole Rund
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - David Brici
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | | | - Alexander M Sykes
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
| | - Annette E Rünker
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Sara Zocher
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Scott Ayton
- The Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Ashley I Bush
- The Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Perry F Bartlett
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Sheng-Tao Hou
- Brain Research Centre and Department of Biology, Southern University of Science and Technology, Shenzhen, China.
| | - Gerd Kempermann
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany.
| | - Tara L Walker
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia; CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany.
| |
Collapse
|
22
|
Al-Amin MM, Sullivan RKP, Alexander S, Carter DA, Bradford D, Burne THJ, Burne THJ. Impaired spatial memory in adult vitamin D deficient BALB/c mice is associated with reductions in spine density, nitric oxide, and neural nitric oxide synthase in the hippocampus. AIMS Neurosci 2022; 9:31-56. [PMID: 35434279 PMCID: PMC8941191 DOI: 10.3934/neuroscience.2022004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/11/2022] [Accepted: 01/20/2022] [Indexed: 02/01/2023] Open
Abstract
Vitamin D deficiency is prevalent in adults and is associated with cognitive impairment. However, the mechanism by which adult vitamin D (AVD) deficiency affects cognitive function remains unclear. We examined spatial memory impairment in AVD-deficient BALB/c mice and its underlying mechanism by measuring spine density, long term potentiation (LTP), nitric oxide (NO), neuronal nitric oxide synthase (nNOS), and endothelial NOS (eNOS) in the hippocampus. Adult male BALB/c mice were fed a control or vitamin D deficient diet for 20 weeks. Spatial memory performance was measured using an active place avoidance (APA) task, where AVD-deficient mice had reduced latency entering the shock zone compared to controls. We characterised hippocampal spine morphology in the CA1 and dentate gyrus (DG) and made electrophysiological recordings in the hippocampus of behaviourally naïve mice to measure LTP. We next measured NO, as well as glutathione, lipid peroxidation and oxidation of protein products and quantified hippocampal immunoreactivity for nNOS and eNOS. Spine morphology analysis revealed a significant reduction in the number of mushroom spines in the CA1 dendrites but not in the DG. There was no effect of diet on LTP. However, hippocampal NO levels were depleted whereas other oxidation markers were unaltered by AVD deficiency. We also showed a reduced nNOS, but not eNOS, immunoreactivity. Finally, vitamin D supplementation for 10 weeks to AVD-deficient mice restored nNOS immunoreactivity to that seen in in control mice. Our results suggest that lower levels of NO and reduced nNOS immunostaining contribute to hippocampal-dependent spatial learning deficits in AVD-deficient mice.
Collapse
Affiliation(s)
- Md. Mamun Al-Amin
- Queensland Brain Institute, The University of Queensland, Brisbane 4072, Australia
| | | | - Suzy Alexander
- Queensland Brain Institute, The University of Queensland, Brisbane 4072, Australia,Queensland Centre for Mental Health Research, Wacol 4076, Australia
| | - David A. Carter
- Queensland Brain Institute, The University of Queensland, Brisbane 4072, Australia
| | - DanaKai Bradford
- Queensland Brain Institute, The University of Queensland, Brisbane 4072, Australia,Australian E-Health Research Centre, CSIRO, Pullenvale 4069, Australia
| | - Thomas H. J. Burne
- Queensland Brain Institute, The University of Queensland, Brisbane 4072, Australia,Queensland Centre for Mental Health Research, Wacol 4076, Australia,* Correspondence: ; Tel: +61 733466371; Fax: +61 733466301
| | | | | | | |
Collapse
|
23
|
Zhou XA, Blackmore DG, Zhuo J, Nasrallah FA, To X, Kurniawan ND, Carlisle A, Vien KY, Chuang KH, Jiang T, Bartlett PF. Neurogenic-dependent changes in hippocampal circuitry underlie the procognitive effect of exercise in aging mice. iScience 2021; 24:103450. [PMID: 34877505 PMCID: PMC8633984 DOI: 10.1016/j.isci.2021.103450] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/22/2021] [Accepted: 11/10/2021] [Indexed: 01/05/2023] Open
Abstract
We have shown that the improvement in hippocampal-based learning in aged mice following physical exercise observed is dependent on neurogenesis in the dentate gyrus (DG) and is regulated by changes in growth hormone levels. The changes in neurocircuitry, however, which may underlie this improvement, remain unclear. Using in vivo multimodal magnetic resonance imaging to track changes in aged mice exposed to exercise, we show the improved spatial learning is due to enhanced DG connectivity, particularly the strengthening of the DG-Cornu Ammonis 3 and the DG-medial entorhinal cortex connections in the dorsal hippocampus. Moreover, we provide evidence that these changes in circuitry are dependent on neurogenesis since they were abrogated by ablation of newborn neurons following exercise. These findings identify the specific changes in hippocampal circuitry that underlie the cognitive improvements resulting from physical activity and show that they are dependent on the activation of neurogenesis in aged animals.
Collapse
Affiliation(s)
- Xiaoqing Alice Zhou
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Daniel G. Blackmore
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Junjie Zhuo
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Fatima A. Nasrallah
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD 4072, Australia
| | - XuanVinh To
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nyoman D. Kurniawan
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Alison Carlisle
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - King-Year Vien
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kai-Hsiang Chuang
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Tianzi Jiang
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Perry F. Bartlett
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
24
|
Blackmore DG, Steyn FJ, Carlisle A, O'Keeffe I, Vien KY, Zhou X, Leiter O, Jhaveri D, Vukovic J, Waters MJ, Bartlett PF. An exercise "sweet spot" reverses cognitive deficits of aging by growth-hormone-induced neurogenesis. iScience 2021; 24:103275. [PMID: 34761193 PMCID: PMC8567379 DOI: 10.1016/j.isci.2021.103275] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/09/2021] [Accepted: 10/12/2021] [Indexed: 11/02/2022] Open
Abstract
Hippocampal function is critical for spatial and contextual learning, and its decline with age contributes to cognitive impairment. Exercise can improve hippocampal function, however, the amount of exercise and mechanisms mediating improvement remain largely unknown. Here, we show exercise reverses learning deficits in aged (24 months) female mice but only when it occurs for a specific duration, with longer or shorter periods proving ineffective. A spike in the levels of growth hormone (GH) and a corresponding increase in neurogenesis during this sweet spot mediate this effect because blocking GH receptor with a competitive antagonist or depleting newborn neurons abrogates the exercise-induced cognitive improvement. Moreover, raising GH levels with GH-releasing hormone agonist improved cognition in nonrunners. We show that GH stimulates neural precursors directly, indicating the link between raised GH and neurogenesis is the basis for the substantially improved learning in aged animals.
Collapse
Affiliation(s)
- Daniel G Blackmore
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Frederik J Steyn
- Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4029, Australia
| | - Alison Carlisle
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Imogen O'Keeffe
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - King-Year Vien
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Xiaoqing Zhou
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Odette Leiter
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Dhanisha Jhaveri
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.,Mater Research Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jana Vukovic
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.,School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Michael J Waters
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Perry F Bartlett
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
25
|
Ribeiro FF, Ferreira F, Rodrigues RS, Soares R, Pedro DM, Duarte-Samartinho M, Aroeira RI, Ferreiro E, Valero J, Solá S, Mira H, Sebastião AM, Xapelli S. Regulation of hippocampal postnatal and adult neurogenesis by adenosine A 2A receptor: Interaction with brain-derived neurotrophic factor. Stem Cells 2021; 39:1362-1381. [PMID: 34043863 DOI: 10.1002/stem.3421] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/06/2021] [Accepted: 05/06/2021] [Indexed: 06/12/2023]
Abstract
Adenosine A2A receptor (A2A R) activation modulates several brain processes, ranging from neuronal maturation to synaptic plasticity. Most of these actions occur through the modulation of the actions of the neurotrophin brain-derived neurotrophic factor (BDNF). In this work, we studied the role of A2A Rs in regulating postnatal and adult neurogenesis in the rat hippocampal dentate gyrus (DG). Here, we show that A2A R activation with CGS 21680 promoted neural stem cell self-renewal, protected committed neuronal cells from cell death and contributed to a higher density of immature and mature neuronal cells, particularly glutamatergic neurons. Moreover, A2A R endogenous activation was found to be essential for BDNF-mediated increase in cell proliferation and neuronal differentiation. Our findings contribute to further understand the role of adenosinergic signaling in the brain and may have an impact in the development of strategies for brain repair under pathological conditions.
Collapse
Affiliation(s)
- Filipa F Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes (iMM, JLB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Filipa Ferreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes (iMM, JLB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Rui S Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes (iMM, JLB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Rita Soares
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes (iMM, JLB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- iMed.ULisboa, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Diogo M Pedro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes (iMM, JLB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Marta Duarte-Samartinho
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes (iMM, JLB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Rita I Aroeira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes (iMM, JLB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Elisabete Ferreiro
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Jorge Valero
- Laboratory of Glial Cell Biology, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Ikerbasque Foundation, Bilbao, Spain
- University of the Basque Country EHU/UPV, Leioa, Spain
| | - Susana Solá
- iMed.ULisboa, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Helena Mira
- Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
- Instituto de Biomedicina de Valencia (IBV-CSIC), Valencia, Spain
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes (iMM, JLB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes (iMM, JLB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
26
|
Valek L, Tegeder I. Failure of Diphtheria Toxin Model to Induce Parkinson-Like Behavior in Mice. Int J Mol Sci 2021; 22:ijms22179496. [PMID: 34502404 PMCID: PMC8430633 DOI: 10.3390/ijms22179496] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/12/2021] [Accepted: 08/29/2021] [Indexed: 12/21/2022] Open
Abstract
Rodent models of Parkinson’s disease are based on transgenic expression of mutant synuclein, deletion of PD genes, injections of MPTP or rotenone, or seeding of synuclein fibrils. The models show histopathologic features of PD such as Lewi bodies but mostly only subtle in vivo manifestations or systemic toxicity. The models only partly mimic a predominant loss of dopaminergic neurons in the substantia nigra. We therefore generated mice that express the transgenic diphtheria toxin receptor (DTR) specifically in DA neurons by crossing DAT-Cre mice with Rosa26 loxP-STOP-loxP DTR mice. After defining a well-tolerated DTx dose, DAT-DTR and DTR-flfl controls were subjected to non-toxic DTx treatment (5 × 100 pg/g) and subsequent histology and behavioral tests. DAT protein levels were reduced in the midbrain, and tyrosine hydroxylase-positive neurons were reduced in the substantia nigra, whereas the pan-neuronal marker NeuN was not affected. Despite the promising histologic results, there was no difference in motor function tests or open field behavior. These are tests in which double mutant Pink1−/−SNCAA53T Parkinson mice show behavioral abnormalities. Higher doses of DTx were toxic in both groups. The data suggest that DTx treatment in mice with Cre/loxP-driven DAT-DTR expression leads to partial ablation of DA-neurons but without PD-reminiscent behavioral correlates.
Collapse
|
27
|
Within-Trial Persistence of Learned Behavior as a Dissociable Behavioral Component in Hippocampus-Dependent Memory Tasks: A Potential Postlearning Role of Immature Neurons in the Adult Dentate Gyrus. eNeuro 2021; 8:ENEURO.0195-21.2021. [PMID: 34281981 PMCID: PMC8387154 DOI: 10.1523/eneuro.0195-21.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 12/20/2022] Open
Abstract
The term “memory strength” generally refers to how well one remembers something. But more precisely it contains multiple modalities, such as how easily, how accurately, how confidently and how vividly we remember it. In human, these modalities of memory strength are dissociable. In this study, we asked whether we can isolate a behavioral component that is dissociable from others in hippocampus-dependent memory tasks in mice, which potentially reflect a modality of memory strength. Using a virus-mediated inducible method, we ablated immature neurons in the dentate gyrus in mice after we trained the mice with hippocampus-dependent memory tasks normally. In memory retrieval tests, these ablated mice initially showed intact performance. However, the ablated mice ceased learned behavior prematurely within a trial compared with control mice. In addition, the ablated mice showed shorter duration of individual episodes of learned behavior. Both affected behavioral measurements point to persistence of learned behavior. Thus, the effect of the postlearning manipulation showed dissociation between initial performance and persistence of learned behavior. These two behavioral components are likely to reflect different brain functions and be mediated by separate mechanisms, which might represent different modalities of memory strength. These simple dissociable measurements in widely used behavioral paradigms would be useful to understand detailed mechanisms underlying the expression of learned behavior and potentially different modalities of memory strength in mice. We also discuss a potential role that immature neurons in the dentate gyrus may play in persistence of learned behavior.
Collapse
|
28
|
Hourigan B, Balay SD, Yee G, Sharma S, Tan Q. Capicua regulates the development of adult-born neurons in the hippocampus. Sci Rep 2021; 11:11725. [PMID: 34083623 PMCID: PMC8175746 DOI: 10.1038/s41598-021-91168-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 05/20/2021] [Indexed: 11/12/2022] Open
Abstract
New neurons continuously arise from neural progenitor cells in the dentate gyrus of the adult hippocampus to support ongoing learning and memory formation. To generate functional adult-born neurons, neural progenitor cells proliferate to expand the precursor cell pool and differentiate into neurons. Newly generated cells then undergo postmitotic maturation to migrate to their final destination and develop elaborate dendritic branching, which allows them to receive input signals. Little is known about factors that regulate neuronal differentiation, migration, and dendrite maturation during adult hippocampal neurogenesis. Here, we show that the transcriptional repressor protein capicua (CIC) exhibits dynamic expression in the adult dentate gyrus. Conditional deletion of Cic from the mouse dentate gyrus compromises the adult neural progenitor cell pool without altering their proliferative potential. We further demonstrate that the loss of Cic impedes neuronal lineage development and disrupts dendritic arborization and migration of adult-born neurons. Our study uncovers a previously unrecognized role of CIC in neurogenesis of the adult dentate gyrus.
Collapse
Affiliation(s)
- Brenna Hourigan
- Department of Cell Biology, University of Alberta, Edmonton, T6J 2H7, Canada
| | - Spencer D Balay
- Department of Cell Biology, University of Alberta, Edmonton, T6J 2H7, Canada.,Research Institute of Molecular Pathology, Vienna Biocenter, Campus-Vienna-Biocenter 1, 1030, Vienna, Austria
| | - Graydon Yee
- Department of Cell Biology, University of Alberta, Edmonton, T6J 2H7, Canada
| | - Saloni Sharma
- Department of Cell Biology, University of Alberta, Edmonton, T6J 2H7, Canada
| | - Qiumin Tan
- Department of Cell Biology, University of Alberta, Edmonton, T6J 2H7, Canada.
| |
Collapse
|
29
|
Selective Ablation of BDNF from Microglia Reveals Novel Roles in Self-Renewal and Hippocampal Neurogenesis. J Neurosci 2021; 41:4172-4186. [PMID: 33785644 DOI: 10.1523/jneurosci.2539-20.2021] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/18/2021] [Accepted: 03/16/2021] [Indexed: 01/22/2023] Open
Abstract
Microglia, the resident immune cells of the CNS, have emerged as key regulators of neural precursor cell activity in the adult brain. However, the microglia-derived factors that mediate these effects remain largely unknown. In the present study, we investigated a role for microglial brain-derived neurotrophic factor (BDNF), a neurotrophic factor with well known effects on neuronal survival and plasticity. Surprisingly, we found that selective genetic ablation of BDNF from microglia increased the production of newborn neurons under both physiological and inflammatory conditions (e.g., LPS-induced infection and traumatic brain injury). Genetic ablation of BDNF from microglia otherwise also interfered with self-renewal/proliferation, reducing their overall density. In conclusion, we identify microglial BDNF as an important factor regulating microglia population dynamics and states, which in turn influences neurogenesis under both homeostatic and pathologic conditions.SIGNIFICANCE STATEMENT (1) Microglial BDNF contributes to self-renewal and density of microglia in the brain. (2) Selective ablation of BDNF in microglia stimulates neural precursor proliferation. (3) Loss of microglial BDNF augments working memory following traumatic brain injury. (4) Benefits of repopulating microglia on brain injury are not mediated via microglial BDNF.
Collapse
|
30
|
Lunardi P, Mansk LMZ, Jaimes LF, Pereira GS. On the novel mechanisms for social memory and the emerging role of neurogenesis. Brain Res Bull 2021; 171:56-66. [PMID: 33753208 DOI: 10.1016/j.brainresbull.2021.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/24/2021] [Accepted: 03/08/2021] [Indexed: 01/25/2023]
Abstract
Social memory (SM) is a key element in social cognition and it encompasses the neural representation of conspecifics, an essential information to guide behavior in a social context. Here we evaluate classical and cutting-edge studies on neurobiology of SM, using as a guiding principle behavioral tasks performed in adult rodents. Our review highlights the relevance of the hippocampus, especially the CA2 region, as a neural substrate for SM and suggest that neural ensembles in the olfactory bulb may also encode SM traces. Compared to other hippocampus-dependent memories, much remains to be done to describe the neurobiological foundations of SM. Nonetheless, we argue that special attention should be paid to neurogenesis. Finally, we pinpoint the remaining open question on whether the hippocampal adult neurogenesis acts through pattern separation to permit the discrimination of highly similar stimuli during behavior.
Collapse
Affiliation(s)
- Paula Lunardi
- Núcleo de Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lara M Z Mansk
- Núcleo de Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Laura F Jaimes
- Núcleo de Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Grace S Pereira
- Núcleo de Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
31
|
Madrid LI, Jimenez-Martin J, Coulson EJ, Jhaveri DJ. Cholinergic regulation of adult hippocampal neurogenesis and hippocampus-dependent functions. Int J Biochem Cell Biol 2021; 134:105969. [PMID: 33727042 DOI: 10.1016/j.biocel.2021.105969] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/10/2021] [Indexed: 10/21/2022]
Abstract
The production and circuit integration of new neurons is one of the defining features of the adult mammalian hippocampus. A wealth of evidence has established that adult hippocampal neurogenesis is exquisitely sensitive to neuronal activity-mediated regulation. How these signals are interpreted and contribute to neurogenesis and hippocampal functions has been a subject of immense interest. In particular, neurotransmitters, in addition to their synaptic roles, have been shown to offer important trophic support. Amongst these, acetylcholine, which has a prominent role in cognition, has been implicated in regulating neurogenesis. In this review, we appraise the evidence linking the contribution of cholinergic signalling to the regulation of adult hippocampal neurogenesis and hippocampus-dependent functions. We discuss open questions that need to be addressed to gain a deeper mechanistic understanding of the role and translational potential of acetylcholine and its receptors in regulating this form of cellular neuroplasticity.
Collapse
Affiliation(s)
- Lidia I Madrid
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Javier Jimenez-Martin
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Elizabeth J Coulson
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Dhanisha J Jhaveri
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia; Mater Research Institute - The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia.
| |
Collapse
|
32
|
Dillingham CM, Milczarek MM, Perry JC, Vann SD. Time to put the mammillothalamic pathway into context. Neurosci Biobehav Rev 2021; 121:60-74. [PMID: 33309908 PMCID: PMC8137464 DOI: 10.1016/j.neubiorev.2020.11.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/10/2020] [Accepted: 11/13/2020] [Indexed: 12/11/2022]
Abstract
The medial diencephalon, in particular the mammillary bodies and anterior thalamic nuclei, has long been linked to memory and amnesia. The mammillary bodies provide a dense input into the anterior thalamic nuclei, via the mammillothalamic tract. In both animal models, and in patients, lesions of the mammillary bodies, mammillothalamic tract and anterior thalamic nuclei all produce severe impairments in temporal and contextual memory, yet it is uncertain why these regions are critical. Mounting evidence from electrophysiological and neural imaging studies suggests that mammillothalamic projections exercise considerable distal influence over thalamo-cortical and hippocampo-cortical interactions. Here, we outline how damage to the mammillary body-anterior thalamic axis, in both patients and animal models, disrupts behavioural performance on tasks that relate to contextual ("where") and temporal ("when") processing. Focusing on the medial mammillary nuclei as a possible 'theta-generator' (through their interconnections with the ventral tegmental nucleus of Gudden) we discuss how the mammillary body-anterior thalamic pathway may contribute to the mechanisms via which the hippocampus and neocortex encode representations of experience.
Collapse
Affiliation(s)
- Christopher M Dillingham
- School of Psychology, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, CF10 3AT, UK
| | - Michal M Milczarek
- School of Psychology, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, CF10 3AT, UK
| | - James C Perry
- School of Psychology, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, CF10 3AT, UK
| | - Seralynne D Vann
- School of Psychology, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, CF10 3AT, UK.
| |
Collapse
|
33
|
Chintamen S, Imessadouene F, Kernie SG. Immune Regulation of Adult Neurogenic Niches in Health and Disease. Front Cell Neurosci 2021; 14:571071. [PMID: 33551746 PMCID: PMC7855589 DOI: 10.3389/fncel.2020.571071] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 12/23/2020] [Indexed: 12/18/2022] Open
Abstract
Microglia regulate neuronal development during embryogenesis, postnatal development, and in specialized microenvironments of the adult brain. Recent evidence demonstrates that in adulthood, microglia secrete factors which modulate adult hippocampal neurogenesis by inhibiting cell proliferation and survival both in vitro and in vivo, maintaining a balance between cell division and cell death in neurogenic niches. These resident immune cells also shape the nervous system by actively pruning synapses during critical periods of learning and engulfing excess neurons. In neurodegenerative diseases, aberrant microglial activity can impede the proper formation and prevent the development of appropriate functional properties of adult born granule cells. Ablating microglia has been presented as a promising therapeutic approach to alleviate the brain of maladaptive immune response. Here, we review key mechanisms through which the immune system actively shapes neurogenic niches throughout the lifespan of the mammalian brain in both health and disease. We discuss how interactions between immune cells and developing neurons may be leveraged for pharmacological intervention and as a means to preserve adult neurogenesis.
Collapse
Affiliation(s)
- Sana Chintamen
- Neurobiology and Behavior, Columbia University Irving Medical Center, New York, NY, United States.,Department of Pediatrics, Columbia University Irving Fefere Medical Center, New York, NY, United States
| | - Fatima Imessadouene
- Department of Pediatrics, Columbia University Irving Fefere Medical Center, New York, NY, United States
| | - Steven G Kernie
- Department of Pediatrics, Columbia University Irving Fefere Medical Center, New York, NY, United States
| |
Collapse
|
34
|
Pléau C, Peret A, Pearlstein E, Scalfati T, Vigier A, Marti G, Michel FJ, Marissal T, Crépel V. Dentate Granule Cells Recruited in the Home Environment Display Distinctive Properties. Front Cell Neurosci 2021; 14:609123. [PMID: 33519383 PMCID: PMC7843370 DOI: 10.3389/fncel.2020.609123] [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: 09/22/2020] [Accepted: 12/23/2020] [Indexed: 11/13/2022] Open
Abstract
The dentate granule cells (DGCs) play a crucial role in learning and memory. Many studies have described the role and physiological properties of these sparsely active neurons using different behavioral contexts. However, the morpho-functional features of DGCs recruited in mice maintained in their home cage (without training), considered as a baseline condition, have not yet been established. Using fosGFP transgenic mice, we observed ex vivo that DGCs recruited in animals maintained in the home cage condition are mature neurons that display a longer dendritic tree and lower excitability compared with non-activated cells. The higher GABAA receptor-mediated shunting inhibition contributes to the lower excitability of DGCs activated in the home environment by shifting the input resistance towards lower values. Remarkably, that shunting inhibition is neither observed in non-activated DGCs nor in DGCs activated during training in virtual reality. In short, our results suggest that strong shunting inhibition and reduced excitability could constitute a distinctive neural signature of mature DGCs recruited in the context of the home environment.
Collapse
Affiliation(s)
- Claire Pléau
- INMED, INSERM UMR1249, Aix-Marseille University, Marseille, France
| | - Angélique Peret
- INMED, INSERM UMR1249, Aix-Marseille University, Marseille, France
| | | | - Thomas Scalfati
- INMED, INSERM UMR1249, Aix-Marseille University, Marseille, France
| | - Alexandre Vigier
- INMED, INSERM UMR1249, Aix-Marseille University, Marseille, France
| | | | | | - Thomas Marissal
- INMED, INSERM UMR1249, Aix-Marseille University, Marseille, France
| | - Valérie Crépel
- INMED, INSERM UMR1249, Aix-Marseille University, Marseille, France
| |
Collapse
|
35
|
Blackmore DG, Turpin F, Palliyaguru T, Evans HT, Chicoteau A, Lee W, Pelekanos M, Nguyen N, Song J, Sullivan RKP, Sah P, Bartlett PF, Götz J. Low-intensity ultrasound restores long-term potentiation and memory in senescent mice through pleiotropic mechanisms including NMDAR signaling. Mol Psychiatry 2021; 26:6975-6991. [PMID: 34040151 PMCID: PMC8760044 DOI: 10.1038/s41380-021-01129-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/31/2021] [Accepted: 04/14/2021] [Indexed: 12/20/2022]
Abstract
Advanced physiological aging is associated with impaired cognitive performance and the inability to induce long-term potentiation (LTP), an electrophysiological correlate of memory. Here, we demonstrate in the physiologically aged, senescent mouse brain that scanning ultrasound combined with microbubbles (SUS+MB), by transiently opening the blood-brain barrier, fully restores LTP induction in the dentate gyrus of the hippocampus. Intriguingly, SUS treatment without microbubbles (SUSonly), i.e., without the uptake of blood-borne factors, proved even more effective, not only restoring LTP, but also ameliorating the spatial learning deficits of the aged mice. This functional improvement is accompanied by an altered milieu of the aged hippocampus, including a lower density of perineuronal nets, increased neurogenesis, and synaptic signaling, which collectively results in improved spatial learning. We therefore conclude that therapeutic ultrasound is a non-invasive, pleiotropic modality that may enhance cognition in elderly humans.
Collapse
Affiliation(s)
- Daniel G. Blackmore
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Fabrice Turpin
- grid.1003.20000 0000 9320 7537Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Tishila Palliyaguru
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Harrison T. Evans
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Antony Chicoteau
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Wendy Lee
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Matthew Pelekanos
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Nghia Nguyen
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Jae Song
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Robert K. P. Sullivan
- grid.1003.20000 0000 9320 7537Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Pankaj Sah
- grid.1003.20000 0000 9320 7537Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia ,grid.263817.90000 0004 1773 1790Joint Center for Neuroscience and Neural Engineering, and Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong Province P. R. China
| | - Perry F. Bartlett
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia ,grid.263817.90000 0004 1773 1790Joint Center for Neuroscience and Neural Engineering, and Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong Province P. R. China
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
36
|
Codd LN, Blackmore DG, Vukovic J, Bartlett PF. Exercise reverses learning deficits induced by hippocampal injury by promoting neurogenesis. Sci Rep 2020; 10:19269. [PMID: 33159114 PMCID: PMC7648755 DOI: 10.1038/s41598-020-76176-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 10/05/2020] [Indexed: 12/18/2022] Open
Abstract
Hippocampal atrophy and cognitive decline are common sequelae of many neurodegenerative disorders, including stroke. To determine whether cognitive decline can be ameliorated by exercise-induced neurogenesis, C57BL/6 mice in which a unilateral hippocampal injury had been induced by injecting the vasoconstrictor endothelin-1 into their right hippocampus, were run voluntarily for 21 days on a running-wheel. We found the severe deficits in spatial learning, as detected by active place-avoidance task, following injury were almost completely restored in animals that ran whereas those that did not run showed no improvement. We show the increase in neurogenesis found in both the injured and contralateral hippocampi following running was responsible for the restoration of learning since bilateral ablation of newborn doublecortin (DCX)-positive neurons abrogated the cognitive improvement, whereas unilateral ablations of DCX-positive neurons did not prevent recovery, demonstrating that elevated neurogenesis in either the damaged or intact hippocampus is sufficient to reverse hippocampal injury-induced deficits.
Collapse
Affiliation(s)
- Lavinia N Codd
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Daniel G Blackmore
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Jana Vukovic
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia.,School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Perry F Bartlett
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
37
|
Arzua T, Yan Y, Jiang C, Logan S, Allison RL, Wells C, Kumar SN, Schäfer R, Bai X. Modeling alcohol-induced neurotoxicity using human induced pluripotent stem cell-derived three-dimensional cerebral organoids. Transl Psychiatry 2020; 10:347. [PMID: 33051447 PMCID: PMC7553959 DOI: 10.1038/s41398-020-01029-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/11/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Maternal alcohol exposure during pregnancy can substantially impact the development of the fetus, causing a range of symptoms, known as fetal alcohol spectrum disorders (FASDs), such as cognitive dysfunction and psychiatric disorders, with the pathophysiology and mechanisms largely unknown. Recently developed human cerebral organoids from induced pluripotent stem cells are similar to fetal brains in the aspects of development and structure. These models allow more relevant in vitro systems to be developed for studying FASDs than animal models. Modeling binge drinking using human cerebral organoids, we sought to quantify the downstream toxic effects of alcohol (ethanol) on neural pathology phenotypes and signaling pathways within the organoids. The results revealed that alcohol exposure resulted in unhealthy organoids at cellular, subcellular, bioenergetic metabolism, and gene expression levels. Alcohol induced apoptosis on organoids. The apoptotic effects of alcohol on the organoids depended on the alcohol concentration and varied between cell types. Specifically, neurons were more vulnerable to alcohol-induced apoptosis than astrocytes. The alcohol-treated organoids exhibit ultrastructural changes such as disruption of mitochondria cristae, decreased intensity of mitochondrial matrix, and disorganized cytoskeleton. Alcohol exposure also resulted in mitochondrial dysfunction and metabolic stress in the organoids as evidenced by (1) decreased mitochondrial oxygen consumption rates being linked to basal respiration, ATP production, proton leak, maximal respiration and spare respiratory capacity, and (2) increase of non-mitochondrial respiration in alcohol-treated organoids compared with control groups. Furthermore, we found that alcohol treatment affected the expression of 199 genes out of 17,195 genes analyzed. Bioinformatic analyses showed the association of these dysregulated genes with 37 pathways related to clinically relevant pathologies such as psychiatric disorders, behavior, nervous system development and function, organismal injury and abnormalities, and cellular development. Notably, 187 of these genes are critically involved in neurodevelopment, and/or implicated in nervous system physiology and neurodegeneration. Furthermore, the identified genes are key regulators of multiple pathways linked in networks. This study extends for the first time animal models of binge drinking-related FASDs to a human model, allowing in-depth analyses of neurotoxicity at tissue, cellular, subcellular, metabolism, and gene levels. Hereby, we provide novel insights into alcohol-induced pathologic phenotypes, cell type-specific vulnerability, and affected signaling pathways and molecular networks, that can contribute to a better understanding of the developmental neurotoxic effects of binge drinking during pregnancy.
Collapse
Affiliation(s)
- Thiago Arzua
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Yasheng Yan
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Congshan Jiang
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Sarah Logan
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Reilly L Allison
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Clive Wells
- Department of Microbiology, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Suresh N Kumar
- Department of Pathology, Children's Research Institute Imaging Core, Neuroscience Imaging Facility, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Richard Schäfer
- Institute for Transfusion Medicine and Immunohaematology, German Red Cross Blood Donor Service Baden-Württemberg-Hessen gGmbH, Goethe University Hospital, 60438, Frankfurt am Main, Germany
| | - Xiaowen Bai
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, 53226, WI, USA.
| |
Collapse
|
38
|
Willis EF, MacDonald KPA, Nguyen QH, Garrido AL, Gillespie ER, Harley SBR, Bartlett PF, Schroder WA, Yates AG, Anthony DC, Rose-John S, Ruitenberg MJ, Vukovic J. Repopulating Microglia Promote Brain Repair in an IL-6-Dependent Manner. Cell 2020; 180:833-846.e16. [PMID: 32142677 DOI: 10.1016/j.cell.2020.02.013] [Citation(s) in RCA: 304] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 11/21/2019] [Accepted: 02/05/2020] [Indexed: 12/20/2022]
Abstract
Cognitive dysfunction and reactive microglia are hallmarks of traumatic brain injury (TBI), yet whether these cells contribute to cognitive deficits and secondary inflammatory pathology remains poorly understood. Here, we show that removal of microglia from the mouse brain has little effect on the outcome of TBI, but inducing the turnover of these cells through either pharmacologic or genetic approaches can yield a neuroprotective microglial phenotype that profoundly aids recovery. The beneficial effects of these repopulating microglia are critically dependent on interleukin-6 (IL-6) trans-signaling via the soluble IL-6 receptor (IL-6R) and robustly support adult neurogenesis, specifically by augmenting the survival of newborn neurons that directly support cognitive function. We conclude that microglia in the mammalian brain can be manipulated to adopt a neuroprotective and pro-regenerative phenotype that can aid repair and alleviate the cognitive deficits arising from brain injury.
Collapse
Affiliation(s)
- Emily F Willis
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Kelli P A MacDonald
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Quan H Nguyen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Adahir Labrador Garrido
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Ellen R Gillespie
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Samuel B R Harley
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Perry F Bartlett
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Wayne A Schroder
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; School of Environment and Science, Griffith University, QLD, Brisbane, Australia
| | - Abi G Yates
- Laboratory of Experimental Neuropathology, Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Daniel C Anthony
- Laboratory of Experimental Neuropathology, Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Stefan Rose-John
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Marc J Ruitenberg
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Jana Vukovic
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
39
|
Canatelli-Mallat M, Lascaray F, Entraigues-Abramson M, Portiansky EL, Blamaceda N, Morel GR, Goya RG. Cryopreservation of a Human Brain and Its Experimental Correlate in Rats. Rejuvenation Res 2020; 23:516-525. [PMID: 32340558 DOI: 10.1089/rej.2019.2245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Several countries have established self-help cryonics groups whose mission is to cryopreserve human bodies or brains after legal death and ship them to cryonics organizations. The objective of this study was to report the first case of human brain cryopreservation in Argentina and complementary experiments in rats. After legal death, the body of a 78-year-old Caucasian woman was transported to a funeral home where her head was submitted to intracarotid perfusion with 5 L cold physiologic saline followed by the same volume of cold saline containing 13% dimethyl sulfoxide and 13% glycerol. The brain was removed, temporarily frozen at -80°C, and shipped to a U.S. cryostasis facility. Three groups of rats were intracardially perfused with fixative but not frozen (Reference group), vitrification solution VM1 (Control group), or the cryoprotection solution used in the patient (Experimental group). Control and Experimental brains were stored at -80°C and subsequently assessed by immunohistochemistry for the adult neuron marker (NeuN), the immature neuron marker doublecortin (DCX), the dopaminergic neuron marker tyrosine hydroxylase, and the presynaptic marker synaptophysin (SYN). The number of NeuN-positive neurons remained unchanged in the experimental brain cortex, whereas the number of immature DCX neurons in the hippocampus fell markedly in the cryoprotected brains. The results were highly variable for hypothalamic dopaminergic neurons. Confocal microscopy for SYN revealed that cryopreservation did not affect the synaptic network in the hippocampus. To our knowledge, this is the first report correlating a human cryoprotection procedure with results in complementary experiments in laboratory animals.
Collapse
Affiliation(s)
| | | | | | - Enrique L Portiansky
- Laboratorio de Análisis de Imágenes (LAI), School of Veterinary Sciences, UNLP, La Plata, Argentina
| | - Néstor Blamaceda
- INIBIOLP-Histology B, Pathology B, School of Medicine, UNLP, La Plata, Argentina
| | - Gustavo R Morel
- INIBIOLP-Histology B, Pathology B, School of Medicine, UNLP, La Plata, Argentina
| | - Rodolfo G Goya
- INIBIOLP-Histology B, Pathology B, School of Medicine, UNLP, La Plata, Argentina
| |
Collapse
|
40
|
Patten KT, González EA, Valenzuela A, Berg E, Wallis C, Garbow JR, Silverman JL, Bein KJ, Wexler AS, Lein PJ. Effects of early life exposure to traffic-related air pollution on brain development in juvenile Sprague-Dawley rats. Transl Psychiatry 2020; 10:166. [PMID: 32483143 PMCID: PMC7264203 DOI: 10.1038/s41398-020-0845-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 12/27/2022] Open
Abstract
Epidemiological studies link traffic-related air pollution (TRAP) to increased risk for various neurodevelopmental disorders (NDDs); however, there are limited preclinical data demonstrating a causal relationship between TRAP and adverse neurodevelopmental outcomes. Moreover, much of the preclinical literature reports effects of concentrated ambient particles or diesel exhaust that do not recapitulate the complexity of real-world TRAP exposures. To assess the developmental neurotoxicity of more realistic TRAP exposures, we exposed male and female rats during gestation and early postnatal development to TRAP drawn directly from a traffic tunnel in Northern California and delivered to animals in real-time. We compared NDD-relevant neuropathological outcomes at postnatal days 51-55 in TRAP-exposed animals versus control subjects exposed to filtered air. As indicated by immunohistochemical analyses, TRAP significantly increased microglial infiltration in the CA1 hippocampus, but decreased astrogliosis in the dentate gyrus. TRAP exposure had no persistent effect on pro-inflammatory cytokine levels in the male or female brain, but did significantly elevate the anti-inflammatory cytokine IL-10 in females. In male rats, TRAP significantly increased hippocampal neurogenesis, while in females, TRAP increased granule cell layer width. TRAP had no effect on apoptosis in either sex. Magnetic resonance imaging revealed that TRAP-exposed females, but not males, also exhibited decreased lateral ventricular volume, which was correlated with increased granule cell layer width in the hippocampus in females. Collectively, these data indicate that exposure to real-world levels of TRAP during gestation and early postnatal development modulate neurodevelopment, corroborating epidemiological evidence of an association between TRAP exposure and increased risk of NDDs.
Collapse
Affiliation(s)
- Kelley T Patten
- Molecular Biosciences, UC Davis School of Veterinary Medicine, Davis, CA, USA
| | - Eduardo A González
- Molecular Biosciences, UC Davis School of Veterinary Medicine, Davis, CA, USA
| | - Anthony Valenzuela
- Molecular Biosciences, UC Davis School of Veterinary Medicine, Davis, CA, USA
| | - Elizabeth Berg
- Psychiatry, UC Davis School of Medicine, Sacramento, CA, USA
| | | | - Joel R Garbow
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Jill L Silverman
- Psychiatry, UC Davis School of Medicine, Sacramento, CA, USA
- The MIND Institute, UC Davis School of Medicine, Sacramento, CA, USA
| | - Keith J Bein
- Air Quality Research Center, UC Davis, Davis, CA, USA
- Center for Health and the Environment, UC Davis, Davis, CA, USA
| | - Anthony S Wexler
- Air Quality Research Center, UC Davis, Davis, CA, USA
- Mechanical and Aerospace Engineering, Civil and Environmental Engineering, and Land, Air and Water Resources, UC Davis, Davis, CA, USA
| | - Pamela J Lein
- Molecular Biosciences, UC Davis School of Veterinary Medicine, Davis, CA, USA.
- The MIND Institute, UC Davis School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
41
|
Dillingham CM, Milczarek MM, Perry JC, Frost BE, Parker GD, Assaf Y, Sengpiel F, O'Mara SM, Vann SD. Mammillothalamic Disconnection Alters Hippocampocortical Oscillatory Activity and Microstructure: Implications for Diencephalic Amnesia. J Neurosci 2019; 39:6696-6713. [PMID: 31235646 PMCID: PMC6703878 DOI: 10.1523/jneurosci.0827-19.2019] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/08/2019] [Accepted: 06/10/2019] [Indexed: 01/13/2023] Open
Abstract
Diencephalic amnesia can be as debilitating as the more commonly known temporal lobe amnesia, yet the precise contribution of diencephalic structures to memory processes remains elusive. Across four cohorts of male rats, we used discrete lesions of the mammillothalamic tract to model aspects of diencephalic amnesia and assessed the impact of these lesions on multiple measures of activity and plasticity within the hippocampus and retrosplenial cortex. Lesions of the mammillothalamic tract had widespread indirect effects on hippocampocortical oscillatory activity within both theta and gamma bands. Both within-region oscillatory activity and cross-regional synchrony were altered. The network changes were state-dependent, displaying different profiles during locomotion and paradoxical sleep. Consistent with the associations between oscillatory activity and plasticity, complementary analyses using several convergent approaches revealed microstructural changes, which appeared to reflect a suppression of learning-induced plasticity in lesioned animals. Together, these combined findings suggest a mechanism by which damage to the medial diencephalon can impact upon learning and memory processes, highlighting an important role for the mammillary bodies in the coordination of hippocampocortical activity.SIGNIFICANCE STATEMENT Information flow within the Papez circuit is critical to memory. Damage to ascending mammillothalamic projections has consistently been linked to amnesia in humans and spatial memory deficits in animal models. Here we report on the changes in hippocampocortical oscillatory dynamics that result from chronic lesions of the mammillothalamic tract and demonstrate, for the first time, that the mammillary bodies, independently of the supramammillary region, contribute to frequency modulation of hippocampocortical theta oscillations. Consistent with the associations between oscillatory activity and plasticity, the lesions also result in a suppression of learning-induced plasticity. Together, these data support new functional models whereby mammillary bodies are important for coordinating hippocampocortical activity rather than simply being a relay of hippocampal information as previously assumed.
Collapse
Affiliation(s)
- Christopher M Dillingham
- School of Psychology, Cardiff University, Cardiff CF10 3AT, United Kingdom
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Michal M Milczarek
- School of Psychology, Cardiff University, Cardiff CF10 3AT, United Kingdom
| | - James C Perry
- School of Psychology, Cardiff University, Cardiff CF10 3AT, United Kingdom
| | - Bethany E Frost
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Greg D Parker
- EMRIC, Cardiff University, Cardiff CF10 3AX, United Kingdom
| | - Yaniv Assaf
- George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel 6997801, and
| | - Frank Sengpiel
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, United Kingdom
| | - Shane M O'Mara
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Seralynne D Vann
- School of Psychology, Cardiff University, Cardiff CF10 3AT, United Kingdom,
| |
Collapse
|
42
|
Shahror RA, Linares GR, Wang Y, Hsueh SC, Wu CC, Chuang DM, Chiang YH, Chen KY. Transplantation of Mesenchymal Stem Cells Overexpressing Fibroblast Growth Factor 21 Facilitates Cognitive Recovery and Enhances Neurogenesis in a Mouse Model of Traumatic Brain Injury. J Neurotrauma 2019; 37:14-26. [PMID: 31298621 DOI: 10.1089/neu.2019.6422] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Traumatic brain injury (TBI) is a progressive and complex pathological condition that results in multiple adverse consequences, including impaired learning and memory. Transplantation of mesenchymal stem cells (MSCs) has produced limited benefits in experimental TBI models. Fibroblast growth factor 21 (FGF21) is a novel metabolic regulator that has neuroprotective effects, promotes remyelination, enhances angiogenesis, and elongates astrocytic processes. In this study, MSCs were genetically engineered to overexpress FGF21 in order to improve their efficacy in TBI. MSCs overexpressing FGF21 (MSC-FGF21) were transplanted to mouse brain by intracerebroventricular injection 24 h after TBI was induced by controlled cortical impact (CCI). Hippocampus-dependent spatial learning and memory, assessed by the Morris water maze test, was markedly decreased 3-4 weeks after TBI, a deficit that was robustly recovered by treatment with MSC-FGF21, but not MSC-mCherry control. Hippocampus-independent learning and memory, assessed by the novel object recognition test, was also impaired; these effects were blocked by treatment with both MSC-FGF21 and MSC-mCherry control. FGF21 protein levels in the ipsilateral hippocampus were drastically reduced 4 weeks post-TBI, a loss that was restored by treatment with MSC-FGF21, but not MSC-mCherry. MSC-FGF21 treatment also partially restored TBI-induced deficits in neurogenesis and maturation of immature hippocampal neurons, whereas MSC-mCherry was less effective. Finally, MSC-FGF21 treatment also normalized TBI-induced impairments in dendritic arborization of hippocampal neurons. Taken together, the results indicate that MSC-FGF21 treatment significantly improved TBI-induced spatial memory deficits, impaired hippocampal neurogenesis, and abnormal dendritic morphology. Future clinical investigations using MSC-FGF21 to improve post-TBI outcomes are warranted.
Collapse
Affiliation(s)
- Rami Ahmad Shahror
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan.,TMU Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan
| | - Gabriel R Linares
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Shih-Chang Hsueh
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan.,TMU Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan
| | - Chung-Che Wu
- TMU Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan.,Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - De-Maw Chuang
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan.,Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Yung-Hsiao Chiang
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan.,TMU Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan.,Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kai-Yun Chen
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan.,TMU Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
43
|
Downregulation of SNX27 expression does not exacerbate amyloidogenesis in the APP/PS1 Alzheimer's disease mouse model. Neurobiol Aging 2019; 77:144-153. [DOI: 10.1016/j.neurobiolaging.2019.01.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/01/2019] [Accepted: 01/13/2019] [Indexed: 12/20/2022]
|
44
|
Adult vitamin D deficiency disrupts hippocampal-dependent learning and structural brain connectivity in BALB/c mice. Brain Struct Funct 2019; 224:1315-1329. [PMID: 30712221 DOI: 10.1007/s00429-019-01840-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 01/22/2019] [Indexed: 12/17/2022]
Abstract
Converging evidence from human and animal studies support an association between vitamin D deficiency and cognitive impairment. Previous studies have shown that hippocampal volume is reduced in adults with vitamin D deficiency as well as in a range of disorders, such as schizophrenia. The aim of the current study was to examine the effect of adult vitamin D (AVD) deficiency on hippocampal-dependent spatial learning, and hippocampal volume and connectivity in healthy adult mice. Ten-week-old male BALB/c mice were fed a control (vitamin D 1500 IU/kg) or vitamin D-depleted (vitamin D 0 IU/kg) diet for a minimum of 10 weeks. The mice were then tested for hippocampal-dependent spatial learning using active place avoidance (APA) and on tests of muscle and motor coordination (rotarod and grip strength). The mice were perfused and brains collected to acquire ex vivo structural and diffusion-weighted images using a 16.4 T MRI scanner. We also performed immunohistochemistry to quantify perineuronal nets (PNNs) and parvalbumin (PV) interneurons in various brain regions. AVD-deficient mice had a lower latency to enter the shock zone on APA, compared to control mice, suggesting impaired hippocampal-dependent spatial learning. There were no differences in rotarod or grip strength, indicating that AVD deficiency did not have an impact on muscle or motor coordination. AVD deficiency did not have an impact on hippocampal volume. However, AVD-deficient mice displayed a disrupted network centred on the right hippocampus with abnormal connectomes among 29 nodes. We found a reduction in PNN positive cells, but no change in PV, centred on the hippocampus. Our results provide compelling evidence to show that AVD deficiency in otherwise healthy adult mice may play a key role in hippocampal-dependent learning and memory formation. We suggest that the spatial learning deficits could be due to the disruption of right hippocampal structural connectivity.
Collapse
|
45
|
Mineyeva OA, Bezriadnov DV, Kedrov AV, Lazutkin AA, Anokhin KV, Enikolopov GN. Radiation Induces Distinct Changes in Defined Subpopulations of Neural Stem and Progenitor Cells in the Adult Hippocampus. Front Neurosci 2019; 12:1013. [PMID: 30686979 PMCID: PMC6333747 DOI: 10.3389/fnins.2018.01013] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 12/17/2018] [Indexed: 11/13/2022] Open
Abstract
While irradiation can effectively treat brain tumors, this therapy also causes cognitive impairments, some of which may stem from the disruption of hippocampal neurogenesis. To study how radiation affects neurogenesis, we combine phenotyping of subpopulations of hippocampal neural stem and progenitor cells with double- and triple S-phase labeling paradigms. Using this approach, we reveal new features of division, survival, and differentiation of neural stem and progenitor cells after exposure to gamma radiation. We show that dividing neural stem cells, while susceptible to damage induced by gamma rays, are less vulnerable than their rapidly amplifying progeny. We also show that dividing stem and progenitor cells that survive irradiation are suppressed in their ability to replicate 0.5–1 day after the radiation exposure. Suppression of division is also observed for cells that entered the cell cycle after irradiation or were not in the S phase at the time of exposure. Determining the longer term effects of irradiation, we found that 2 months after exposure, radiation-induced suppression of division is partially relieved for both stem and progenitor cells, without evidence for compensatory symmetric divisions as a means to restore the normal level of neurogenesis. By that time, most mature young neurons, born 2–4 weeks after the irradiation, still bear the consequences of radiation exposure, unlike younger neurons undergoing early stages of differentiation without overt signs of deficient maturation. Later, 6 months after an exposure to 5 Gy, cell proliferation and neurogenesis are further impaired, though neural stem cells are still available in the niche, and their pool is preserved. Our results indicate that various subpopulations of stem and progenitor cells in the adult hippocampus have different susceptibility to gamma radiation, and that neurogenesis, even after a temporary restoration, is impaired in the long term after exposure to gamma rays. Our study provides a framework for investigating critical issues of neural stem cell maintenance, aging, interaction with their microenvironment, and post-irradiation therapy.
Collapse
Affiliation(s)
- Olga A Mineyeva
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,P. K. Anokhin Research Institute of Normal Physiology, Moscow, Russia.,National Research Center "Kurchatov Institute," Moscow, Russia
| | - Dmitri V Bezriadnov
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,P. K. Anokhin Research Institute of Normal Physiology, Moscow, Russia
| | - Alexander V Kedrov
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,P. K. Anokhin Research Institute of Normal Physiology, Moscow, Russia
| | - Alexander A Lazutkin
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,P. K. Anokhin Research Institute of Normal Physiology, Moscow, Russia.,N.N. Burdenko Neurosurgery Institute, Moscow, Russia
| | - Konstantin V Anokhin
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,P. K. Anokhin Research Institute of Normal Physiology, Moscow, Russia.,National Research Center "Kurchatov Institute," Moscow, Russia
| | - Grigori N Enikolopov
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,Center for Developmental Genetics, Department of Anesthesiology, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
46
|
Blackmore DG, Turpin F, Mohamed AZ, Zong F, Pandit R, Pelekanos M, Nasrallah F, Sah P, Bartlett PF, Götz J. Multimodal analysis of aged wild-type mice exposed to repeated scanning ultrasound treatments demonstrates long-term safety. Am J Cancer Res 2018; 8:6233-6247. [PMID: 30613294 PMCID: PMC6299703 DOI: 10.7150/thno.27941] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 10/30/2018] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier presents a major challenge for the delivery of therapeutic agents to the brain; however, it can be transiently opened by combining low intensity ultrasound with microbubble infusion. Studies evaluating this technology have largely been performed in rodents, including models of neurological conditions. However, despite promising outcomes in terms of drug delivery and the amelioration of neurological impairments, the potential for long-term adverse effects presents a major concern in the context of clinical applications. Methods: To fill this gap, we repeatedly treated 12-month-old wild-type mice with ultrasound, followed by a multimodal analysis for up to 18 months of age. Results: We found that spatial memory in these aged mice was not adversely affected as assessed in the active place avoidance test. Sholl analysis of Golgi impregnations in the dentate gyrus of the hippocampus did not reveal any changes to the neuronal cytoarchitecture. Long-term potentiation, a cellular correlate of memory, was still achievable, magnetic resonance spectroscopy revealed no major changes in metabolites, and diffusion tensor imaging revealed normal microstructure and tissue integrity in the hippocampus. More specifically, all measures of diffusion appeared to support a neuroprotective effect of ultrasound treatment on the brain. Conclusion: This multimodal analysis indicates that therapeutic ultrasound for blood-brain barrier opening is safe and potentially protective in the long-term, underscoring its validity as a potential treatment modality for diseases of the brain.
Collapse
|
47
|
Qian L, Milne MR, Shepheard S, Rogers ML, Medeiros R, Coulson EJ. Removal of p75 Neurotrophin Receptor Expression from Cholinergic Basal Forebrain Neurons Reduces Amyloid-β Plaque Deposition and Cognitive Impairment in Aged APP/PS1 Mice. Mol Neurobiol 2018; 56:4639-4652. [DOI: 10.1007/s12035-018-1404-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 10/22/2018] [Indexed: 12/14/2022]
|
48
|
Zalucki O, Harkins D, Harris L, Burne THJ, Gronostajski RM, Piper M. Analysis of hippocampal-dependent learning and memory behaviour in mice lacking Nfix from adult neural stem cells. BMC Res Notes 2018; 11:564. [PMID: 30081965 PMCID: PMC6080370 DOI: 10.1186/s13104-018-3652-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 07/25/2018] [Indexed: 11/16/2022] Open
Abstract
Objective The active place avoidance task (APA) is a behavioural task used to assess learning and memory in rodents. This task relies on the hippocampus, a region of the cerebral cortex capable of generating new neurons from neural stem cells. In this study, to gain further insight into the behavioural phenotype of mice deficient in the transcription factor Nfix, a gene expressed by adult neural stem cells, we examined learning and memory parameters from the APA task that were not published in our original investigation. We analysed time to first and second shock, maximum path and time of shock avoidance, number of entries into the shock zone and time spent in the shock zone. We also assessed performance in the APA task based on sex. Results We found mice deficient in Nfix displayed decreased latency to second shock compared to the control mice. Nfix deficient mice entered the shock zone more frequently and also spent more time in the shock zone. Our data provides further insights into the memory deficits evident in Nfix mutant mice, indicating these mice have a memory retrieval problem and may employ a different navigation strategy in the APA task. Electronic supplementary material The online version of this article (10.1186/s13104-018-3652-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Oressia Zalucki
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Danyon Harkins
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Lachlan Harris
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Thomas H J Burne
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, Australia.,Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD, 4076, Australia
| | - Richard M Gronostajski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Michael Piper
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia. .,Queensland Brain Institute, The University of Queensland, Brisbane, 4072, Australia.
| |
Collapse
|
49
|
Newly Generated Cells in the Dentate Gyrus Differentially Respond to Brief Spatial Exploration and Forced Swim in Adult Female Balb/C Mice. Neural Plast 2018; 2018:4960869. [PMID: 29951090 PMCID: PMC5987312 DOI: 10.1155/2018/4960869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/29/2018] [Accepted: 05/02/2018] [Indexed: 12/31/2022] Open
Abstract
Neurogenesis in the hippocampus is influenced by several factors including external stimuli. In addition to their involvement in learning and memory processes, newborn neurons of the dentate gyrus (DG) buffer against the effects of stress. Although the response of these cells to environmental stimuli has been shown, the age of the cells that respond to a brief spatial exploration or a stressful situation produced by forced-swim stress in adult female Balb/C mice is still unknown. Here, we investigated the activation of newborn neurons after three (IdU) or six weeks (CldU) postlabelling with the expression of Arc in the same mice but exposed to different environmental stimuli. Mice housed in standard conditions showed an increase in the activation of CldU-labelled cells after two exposures to a brief spatial exploration but no increase in the activation of IdU-labelled cells compared with the control group. Additionally, we analysed neuronal activation in the DG of mice housed in standard conditions and further exposed to forced-swim stress. We found a decreased activation of IdU-labelled cells in mice exposed to forced-swim stress with increase number of CldU-labelled cells. Our results suggest that based on their time postlabelling, newly generated hippocampal neurons show a different response to several environmental stimuli.
Collapse
|
50
|
A behavioral task with more opportunities for memory acquisition promotes the survival of new neurons in the adult dentate gyrus. Sci Rep 2018; 8:7369. [PMID: 29743494 PMCID: PMC5943398 DOI: 10.1038/s41598-018-25331-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/29/2018] [Indexed: 12/14/2022] Open
Abstract
It has been suggested that the dentate gyrus, particularly its new neurons generated via adult neurogenesis, is involved in memory acquisition and recall. Here, we trained rats in two types of Morris water maze tasks that are differentially associated with these two memory processes, and examined whether new neurons are differently affected by the two tasks performed during the second week of neuronal birth. Our results indicate that the task involving more opportunities to acquire new information better supports the survival of new neurons. Further, we assessed whether the two tasks differentially induce the expression of an immediate early gene, Zif268, which is known to be induced by neuronal activation. While the two tasks differentially induce Zif268 expression in the dentate gyrus, the proportions of new neurons activated were similar between the two tasks. Thus, we conclude that while the two tasks differentially activate the dentate gyrus, the task involving more opportunities for memory acquisition during the second week of the birth of new neurons better promotes the survival of the new neurons.
Collapse
|