451
|
Kang H, Zhou H, Ye Y, Yang J, Liu Z, He P, Li B, Wu Y, Wang Y, Tu Y. Tieguanyin Oolong Tea Extracts Alleviate Behavioral Abnormalities by Modulating Neuroinflammation in APP/PS1 Mouse Model of Alzheimer's Disease. Foods 2021; 11:81. [PMID: 35010207 PMCID: PMC8750439 DOI: 10.3390/foods11010081] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease; tea components have important neuroprotective effects. This article explores the effects and mechanisms of Qingxiang Tiguanyin (Tgy-Q), Nongxiang Tieguanyin (Tgy-N), and Chenxiang Tieguanyin (Tgy-C) extracts on APP/PS1 AD model mice. Morris water maze and new object recognition experiments show that Tieguanyin extracts can effectively enhance the cognitive ability of APP/PS1 mice. H&E staining, Nissl staining, and immunohistochemical staining show that Tieguanyin extracts make nerve cell boundaries and nucleoli become clearer, relieve nucleus pyknosis, and effectively reduce Aβ1-40 and Aβ1-42 in the hippocampus and cortex. They also restore the morphology of microglia and astrocytes. In addition, Tieguanyin extracts can balance the oxidative stress level in the brain of APP/PS1 mice by improving the antioxidant capacity. Western blot results show that Tieguanyin extracts can reduce the expression of NF-κB p65, TNF-α, IL-1β, IL-6, COX-2, and iNOS in mouse brain, which demonstrates that Tieguanyin extracts improves cognitive ability by alleviating inflammation. This article demonstrates for the first time that Tieguanyin extracts can inhibit the excessive activation of the NF-κB p65 signaling pathway and improve the antioxidant capacity in the cerebral cortex and hippocampus, to improve the cognitive ability of APP/PS1 mice. Our results shed light into the beneficial of Tieguanyin tea extracts on preventing and alleviating AD diseases.
Collapse
Affiliation(s)
- Hyunuk Kang
- Department of Tea Science, Zhejiang University, Hangzhou 310058, China; (H.K.); (H.Z.); (Y.Y.); (Z.L.); (P.H.); (B.L.); (Y.W.)
| | - Hui Zhou
- Department of Tea Science, Zhejiang University, Hangzhou 310058, China; (H.K.); (H.Z.); (Y.Y.); (Z.L.); (P.H.); (B.L.); (Y.W.)
| | - Yushan Ye
- Department of Tea Science, Zhejiang University, Hangzhou 310058, China; (H.K.); (H.Z.); (Y.Y.); (Z.L.); (P.H.); (B.L.); (Y.W.)
| | - Jiangfan Yang
- Key Laboratory of Tea Science, College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China;
| | - Zhonghua Liu
- Department of Tea Science, Zhejiang University, Hangzhou 310058, China; (H.K.); (H.Z.); (Y.Y.); (Z.L.); (P.H.); (B.L.); (Y.W.)
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China
| | - Puming He
- Department of Tea Science, Zhejiang University, Hangzhou 310058, China; (H.K.); (H.Z.); (Y.Y.); (Z.L.); (P.H.); (B.L.); (Y.W.)
| | - Bo Li
- Department of Tea Science, Zhejiang University, Hangzhou 310058, China; (H.K.); (H.Z.); (Y.Y.); (Z.L.); (P.H.); (B.L.); (Y.W.)
| | - Yuanyuan Wu
- Department of Tea Science, Zhejiang University, Hangzhou 310058, China; (H.K.); (H.Z.); (Y.Y.); (Z.L.); (P.H.); (B.L.); (Y.W.)
| | - Yaomin Wang
- Key Laboratory of Horticulture Plant Biology, Ministry of Education, College of Horticulture and Forestry Sciences, Huazhong Agricultural University, Wuhan 430070, China
| | - Youying Tu
- Department of Tea Science, Zhejiang University, Hangzhou 310058, China; (H.K.); (H.Z.); (Y.Y.); (Z.L.); (P.H.); (B.L.); (Y.W.)
| |
Collapse
|
452
|
Kelty TJ, Mao X, Kerr NR, Childs TE, Ruegsegger GN, Booth FW. Resistance-exercise training attenuates LPS-induced astrocyte remodeling and neuroinflammatory cytokine expression in female Wistar rats. J Appl Physiol (1985) 2021; 132:317-326. [PMID: 34941437 DOI: 10.1152/japplphysiol.00571.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neuroinflammation is an early detectable marker of mild cognitive impairment, the transition state between normal cognition and dementia. Resistance-exercise training can attenuate the cognitive decline observed in patients with mild cognitive impairment. However, the underlying mechanisms of resistance training effects are largely unknown. To further elucidate mechanisms of the known cognitive health benefits from resistance-exercise training, we tested if three weeks of resistance-exercise training could ameliorate lipopolysaccharide-induced neuroinflammation. Five-week-old female Wistar rats received intracerebroventricular injections of lipopolysaccharides to induce neuroinflammation and cognitive impairment. Rats then underwent three weeks of progressive ladder climbing to recapitulate resistance-exercise training in humans. Cognition was assessed towards the end of the training period by novelty object recognition testing. Neuroinflammation was measured one and 24-hours after the last resistance-exercise training workout. Resistance-exercise training ameliorated cognitive impairment, diminished lipopolysaccharide-induced neuroinflammatory cytokine expression, and attenuated astrocyte remodeling in the dentate gyrus 24-hours post exercise. Here, we provide evidence that the ladder-climbing model of resistance-exercise training in rats can improve cognition as early as three weeks. Additionally, these data support the hypothesis that resistance exercise can reduce lipopolysaccharide-induced neuroinflammation in the dentate gyrus.
Collapse
Affiliation(s)
- Taylor J Kelty
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
| | - Xuansong Mao
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
| | - Nathan R Kerr
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
| | - Thomas E Childs
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
| | - Gregory N Ruegsegger
- Department of Health and Human Performance, University of Wisconsin-River Falls, River Falls, WI, United States
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| |
Collapse
|
453
|
Tropea MR, Sanfilippo G, Giannino F, Davì V, Gulisano W, Puzzo D. Innate Preferences Affect Results of Object Recognition Task in Wild Type and Alzheimer's Disease Mouse Models. J Alzheimers Dis 2021; 85:1343-1356. [PMID: 34924388 PMCID: PMC8925114 DOI: 10.3233/jad-215209] [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] [Indexed: 11/15/2022]
Abstract
BACKGROUND Object recognition task (ORT) is a widely used behavioral paradigm to assess memory in rodent models, due to its easy technical execution, the lack of aversive stressful stimuli, and the possibility to repeat the test on the same animals. However, mouse exploration might be strongly influenced by a variety of variables. OBJECTIVE To study whether innate preferences influenced exploration in male and female wild type mice and the Alzheimer's disease (AD) model 3xTg. METHODS We first evaluated how object characteristics (material, size, and shape) influence exploration levels, latency, and exploration modality. Based on these findings, we evaluated whether these innate preferences biased the results of ORT performed in wild type mice and AD models. RESULTS Assessment of Exploration levels, i.e., the time spent in exploring a certain object in respect to the total exploration time, revealed an innate preference for objects made in shiny materials, such as metal and glass. A preference for bigger objects characterized by higher affordance was also evident, especially in male mice. When performing ORT, exploration was highly influenced by these innate preferences. Indeed, both wild type and AD mice spent more time in exploring the metal object, regardless of its novelty. Furthermore, the use of objects with higher affordance such as the cube was a confounding factor leading to "false" results that distorted ORT interpretation. CONCLUSION When designing exploration-based behavioral experiments aimed at assessing memory in healthy and AD mice, object characteristics should be carefully evaluated to improve scientific outcomes and minimize possible biases.
Collapse
Affiliation(s)
- Maria Rosaria Tropea
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Giulia Sanfilippo
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Federico Giannino
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Valentina Davì
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Walter Gulisano
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Daniela Puzzo
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy.,Oasi Research Institute-IRCCS, Troina, Italy
| |
Collapse
|
454
|
Bodden C, Pang TY, Feng Y, Mridha F, Kong G, Li S, Watt MJ, Reichelt AC, Hannan AJ. Intergenerational effects of a paternal Western diet during adolescence on offspring gut microbiota, stress reactivity, and social behavior. FASEB J 2021; 36:e21981. [PMID: 34907601 DOI: 10.1096/fj.202100920rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/16/2021] [Accepted: 09/22/2021] [Indexed: 12/28/2022]
Abstract
The global consumption of highly processed, calorie-dense foods has contributed to an epidemic of overweight and obesity, along with negative consequences for metabolic dysfunction and disease susceptibility. As it becomes apparent that overweight and obesity have ripple effects through generations, understanding of the processes involved is required, in both maternal and paternal epigenetic inheritance. We focused on the patrilineal effects of a Western-style high-fat (21%) and high-sugar (34%) diet (WD) compared to control diet (CD) during adolescence and investigated F0 and F1 mice for physiological and behavioral changes. F0 males (fathers) showed increased body weight, impaired glycemic control, and decreased attractiveness to females. Paternal WD caused significant phenotypic changes in F1 offspring, including higher body weights of pups, increased Actinobacteria abundance in the gut microbiota (ascertained using 16S microbiome profiling), a food preference for WD pellets, increased male dominance and attractiveness to females, as well as decreased behavioral despair. These results collectively demonstrate the long-term intergenerational effects of a Western-style diet during paternal adolescence. The behavioral and physiological alterations in F1 offspring provide evidence of adaptive paternal programming via epigenetic inheritance. These findings have important implications for understanding paternally mediated intergenerational inheritance, and its relevance to offspring health and disease susceptibility.
Collapse
Affiliation(s)
- Carina Bodden
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Terence Y Pang
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Yingshi Feng
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Faria Mridha
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Geraldine Kong
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Shanshan Li
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Matthew J Watt
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Amy C Reichelt
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia.,Department of Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Anthony J Hannan
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia.,Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
455
|
Long-term caffeine treatment of Alzheimer mouse models ameliorates behavioural deficits and neuron loss and promotes cellular and molecular markers of neurogenesis. Cell Mol Life Sci 2021; 79:55. [PMID: 34913091 PMCID: PMC8738505 DOI: 10.1007/s00018-021-04062-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/15/2021] [Accepted: 11/24/2021] [Indexed: 01/04/2023]
Abstract
Epidemiological studies indicate that the consumption of caffeine, the most commonly ingested psychoactive substance found in coffee, tea or soft drinks, reduces the risk of developing Alzheimer’s disease (AD). Previous treatment studies with transgenic AD mouse models reported a reduced amyloid plaque load and an amelioration of behavioral deficits. It has been further shown that moderate doses of caffeine have the potential to attenuate the health burden in preclinical mouse models of a variety of brain disorders (reviewed in Cunha in J Neurochem 139:1019–1055, 2016). In the current study, we assessed whether long-term caffeine consumption affected hippocampal neuron loss and associated behavioral deficits in the Tg4-42 mouse model of AD. Treatment over a 4-month period reduced hippocampal neuron loss, rescued learning and memory deficits, and ameliorated impaired neurogenesis. Neuron-specific RNA sequencing analysis in the hippocampus revealed an altered expression profile distinguished by the up-regulation of genes linked to synaptic function and processes, and to neural progenitor proliferation. Treatment of 5xFAD mice, which develop prominent amyloid pathology, with the same paradigm also rescued behavioral deficits but did not affect extracellular amyloid-β (Aβ) levels or amyloid precursor protein (APP) processing. These findings challenge previous assumptions that caffeine is anti-amyloidogenic and indicate that the promotion of neurogenesis might play a role in its beneficial effects.
Collapse
|
456
|
Altered dorsal functional connectivity after post-weaning social isolation and resocialization in mice. Neuroimage 2021; 245:118740. [PMID: 34808365 DOI: 10.1016/j.neuroimage.2021.118740] [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: 07/13/2021] [Revised: 10/27/2021] [Accepted: 11/16/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Social isolation (SI) leads to various mental health disorders. Despite abundant studies on behavioral and neurobiological changes induced by post-weaning SI, the characterization of its imaging correlates, such as resting-state functional connectivity (RSFC), is critically lacking. In addition, the effects of resocialization after isolation remain unclear. Therefore, this study aimed to explore the effects of 1) SI on cortical functional connectivity and 2) subsequent resocialization on behavior and functional connectivity. METHODS Behavioral tests were conducted to validate the post-weaning SI mouse model, which is isolated during the juvenile period. Wide-field optical mapping was performed to observe both neuronal and hemodynamic signals in the cortex under anesthesia. Using seed-based and graph theoretical analyses, RSFC was analyzed. SI mice were then resocialized and the array of behavior and imaging tests was conducted. RESULTS Behaviorally, SI mice showed elevated anxiety, social preference, and aggression. RSFC analyses using the seed-based approach revealed decreased cortical functional connectivity in SI mice, especially in the frontal region. Graph network analyses demonstrated significant reduction in network segregation measures. After resocialization, mice exhibited recovered anxiogenic and aggressive behavior, but RSFC data did not show significant changes. CONCLUSIONS We observed an overall decrease in functional connectivity in SI mice. Moreover, resocialization restored the disruptions in behavioral patterns but functional connectivity was not recovered. To our knowledge, this is the first study to report that, despite the recovering tendencies of behavior in resocialized mice, similar changes in RSFC were not observed. This suggests that disruptions in functional connectivity caused by social isolation remain as long-term sequelae.
Collapse
|
457
|
Han J, Hyun J, Park J, Jung S, Oh Y, Kim Y, Ryu SH, Kim SH, Jeong EI, Jo DG, Park SH, Jung YK. Aberrant role of pyruvate kinase M2 in the regulation of gamma-secretase and memory deficits in Alzheimer's disease. Cell Rep 2021; 37:110102. [PMID: 34879266 DOI: 10.1016/j.celrep.2021.110102] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 09/25/2021] [Accepted: 11/15/2021] [Indexed: 10/19/2022] Open
Abstract
Toxic amyloid beta (Aβ) species cause synaptic dysfunction and neurotoxicity in Alzheimer's disease (AD). As of yet, however, there are no reported regulators for gamma-secretase, which links a risky environment to amyloid accumulation in AD. Here, we report that pyruvate kinase M2 (PKM2) is a positive regulator of gamma-secretase under hypoxia. From a genome-wide functional screen, we identify PKM2 as a gamma-secretase activator that is highly expressed in the brains of both patients and murine models with AD. PKM2 regulates Aβ production and the amount of active gamma-secretase complex by changing the gene expression of aph-1 homolog. Hypoxia induces PKM2 expression, thereby promoting gamma-secretase activity. Moreover, transgenic expression of PKM2 in 3xTg AD model mice enhances hippocampal production of Aβ and exacerbates the impairment of spatial and recognition memory. Taken together, these findings indicate that PKM2 is an important gamma-secretase regulator that promotes Aβ production and memory impairment under hypoxia.
Collapse
Affiliation(s)
- Jonghee Han
- School of Biological Science, Seoul National University, Seoul 08826, Korea; Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Junho Hyun
- School of Biological Science, Seoul National University, Seoul 08826, Korea
| | - Jaesang Park
- School of Biological Science, Seoul National University, Seoul 08826, Korea
| | - Sunmin Jung
- School of Biological Science, Seoul National University, Seoul 08826, Korea
| | - Yoonseo Oh
- School of Biological Science, Seoul National University, Seoul 08826, Korea
| | - Youbin Kim
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul 08826, Korea
| | - Shin-Hyeon Ryu
- School of Biological Science, Seoul National University, Seoul 08826, Korea
| | - Seo-Hyun Kim
- School of Biological Science, Seoul National University, Seoul 08826, Korea
| | - Eun Il Jeong
- School of Biological Science, Seoul National University, Seoul 08826, Korea
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Gyunggi-do 16419, Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Yong-Keun Jung
- School of Biological Science, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
458
|
Su Y, Lian J, Hodgson J, Zhang W, Deng C. Prenatal Poly I:C Challenge Affects Behaviors and Neurotransmission via Elevated Neuroinflammation Responses in Female Juvenile Rats. Int J Neuropsychopharmacol 2021; 25:160-171. [PMID: 34893855 PMCID: PMC8832231 DOI: 10.1093/ijnp/pyab087] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/12/2021] [Accepted: 12/04/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Exposure to polyriboinosinic-polyribocytidylic acid (Poly I:C) in pregnant rats has been reported to cause schizophrenia-like behaviors and abnormal neurotransmissions in adult, particularly male, offspring. However, what is less well understood are the effects of maternal Poly I:C exposure on adolescent behaviors and neurotransmission in female juvenile rats. METHODS Female adolescent Poly I:C offspring were constructed by treating with 5 mg/kg Poly I:C on timed pregnant rats (gestation day 15). A battery of behavioral tests was conducted during postnatal day 35-60. Neurotransmitter receptors and inflammation markers in brain regions were evaluated by RT-qPCR on postnatal day 60. RESULTS Open field, elevated plus maze, and forced swimming tests revealed that prenatal Poly I:C exposure led to elevated anxiety-like and depression-like behaviors in female adolescent offspring. Deficits in pre-pulse inhibition and social interaction were also observed. However, the Poly I:C rats had better performance than the controls in the novel object recognition memory test, which demonstrated a behavioral phenotype with improved cognitive function. Prenatal Poly I:C exposure caused brain region-specific elevation of the P2X7 receptor- and NF-κB-NLRP3-IL-1β inflammatory signaling in female juvenile rats. Prenatal Poly I:C exposure decreased expression of GABAA receptor subunits Gabrb3 in the prefrontal cortex and Gabrb1 and dopamine D2 receptor in the hippocampus, but increased NMDA receptor subunit Grin2a in the prefrontal cortex, 5-HT2A in the hippocampus, and Gabrb3 and D2 receptor in the nucleus accumben. CONCLUSIONS Prenatal Poly I:C challenge causes behavioral deficits and brain-specific neurotransmission changes via elevated neuroinflammation responses in female adolescent offspring rats.
Collapse
Affiliation(s)
- Yueqing Su
- The School of Public Health, Fujian Medical University, Fuzhou, China,Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China,Antipsychotic Research Laboratory, Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia,School of Medicine, and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Jiamei Lian
- School of Medicine, and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - James Hodgson
- Antipsychotic Research Laboratory, Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia,School of Medicine, and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Wenchang Zhang
- The School of Public Health, Fujian Medical University, Fuzhou, China
| | - Chao Deng
- Antipsychotic Research Laboratory, Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia,School of Medicine, and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia,Correspondence: Chao Deng, PhD, Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia ()
| |
Collapse
|
459
|
Loureiro-Campos E, Mateus-Pinheiro A, Patrício P, Soares-Cunha C, Silva J, Sardinha VM, Mendes-Pinheiro B, Silveira-Rosa T, Domingues AV, Rodrigues AJ, Oliveira J, Sousa N, Alves ND, Pinto L. Constitutive deficiency of the neurogenic hippocampal modulator AP2γ promotes anxiety-like behavior and cumulative memory deficits in mice from juvenile to adult periods. eLife 2021; 10:70685. [PMID: 34859784 PMCID: PMC8709574 DOI: 10.7554/elife.70685] [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: 05/26/2021] [Accepted: 12/02/2021] [Indexed: 11/23/2022] Open
Abstract
The transcription factor activating protein two gamma (AP2γ) is an important regulator of neurogenesis both during embryonic development as well as in the postnatal brain, but its role for neurophysiology and behavior at distinct postnatal periods is still unclear. In this work, we explored the neurogenic, behavioral, and functional impact of a constitutive and heterozygous AP2γ deletion in mice from early postnatal development until adulthood. AP2γ deficiency promotes downregulation of hippocampal glutamatergic neurogenesis, altering the ontogeny of emotional and memory behaviors associated with hippocampus formation. The impairments induced by AP2γ constitutive deletion since early development leads to an anxious-like phenotype and memory impairments as early as the juvenile phase. These behavioral impairments either persist from the juvenile phase to adulthood or emerge in adult mice with deficits in behavioral flexibility and object location recognition. Collectively, we observed a progressive and cumulative impact of constitutive AP2γ deficiency on the hippocampal glutamatergic neurogenic process, as well as alterations on limbic-cortical connectivity, together with functional behavioral impairments. The results herein presented demonstrate the modulatory role exerted by the AP2γ transcription factor and the relevance of hippocampal neurogenesis in the development of emotional states and memory processes.
Collapse
Affiliation(s)
- Eduardo Loureiro-Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Guimarães, Portugal
| | - António Mateus-Pinheiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Guimarães, Portugal
| | - Patrícia Patrício
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Guimarães, Portugal
| | - Carina Soares-Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Guimarães, Portugal
| | - Joana Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Guimarães, Portugal
| | - Vanessa Morais Sardinha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Guimarães, Portugal
| | - Bárbara Mendes-Pinheiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Guimarães, Portugal
| | - Tiago Silveira-Rosa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Guimarães, Portugal
| | - Ana Verónica Domingues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Guimarães, Portugal
| | - Ana João Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
| | - João Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Guimarães, Portugal.,IPCA-EST-2Ai, Polytechnic Institute of Cávado and Ave, Applied Artificial Intelligence Laboratory, Campus of IPCA, Barcelos, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Guimarães, Portugal
| | - Nuno Dinis Alves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Guimarães, Portugal
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Guimarães, Portugal
| |
Collapse
|
460
|
Özkan E, Çetin-Taş Y, Şekerdağ E, Kızılırmak AB, Taş A, Yıldız E, Yapıcı-Eser H, Karahüseyinoğlu S, Zeybel M, Gürsoy-Özdemir Y. Blood-brain barrier leakage and perivascular collagen accumulation precede microvessel rarefaction and memory impairment in a chronic hypertension animal model. Metab Brain Dis 2021; 36:2553-2566. [PMID: 34118020 DOI: 10.1007/s11011-021-00767-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 06/06/2021] [Indexed: 01/07/2023]
Abstract
Hypertension (HT) is one of the main causes of vascular dementia, lead to cognitive decline. Here, we investigated the relationship between cerebral microvessels, pericytes, extracellular matrix (ECM) accumulation, blood-brain barrier (BBB) breakdown, and memory impairment at mid-life in a chronic hypertension animal model. Spontaneously hypertensive rats (SHRs) (n = 20) are chosen for the model and age matched Wistar rats (n = 16) as controls. Changes in brain microvasculature and in vitro experiments are shown with immunofluorescence studies and cognition with open field, novel object recognition, and Y maze tests. There was a significant reduction in pericyte coverage in SHRs (p = 0.021), while the quantitative parameters of the cerebral microvascular network were not different between groups. On the other hand, parenchymal albumin leakage, as a Blood-brain barrier (BBB) breakdown marker, was prominent in SHRs (p = 0.023). Extracellular matrix (ECM) components, collagen type 1, 3 and 4 were significantly increased (accumulated) around microvasculature in SHRs (p = 0.011, p = 0.013, p = 0.037, respectively). Furthermore, in vitro experiments demonstrated that human brain vascular pericytes but not astrocytes and endothelial cells secreted type I collagen upon TGFβ1 exposure pointing out a possible role of pericytes in increased collagen accumulation around cerebral microvasculature due to HT. Furthermore, valsartan treatment decreased the amount of collagen type 1 secreted by pericytes after TGFβ1 exposure. At the time of evaluation, SHRs did not demonstrate cognitive decline and memory impairments. Our results showed that chronic HT causes ECM accumulation and BBB leakage before leading to memory impairments and therefore, pericytes could be a novel target for preventing vascular dementia.
Collapse
Affiliation(s)
- Esra Özkan
- Koç University Research Center for Translational Medicine, Istanbul, Turkey.
- Koç University Hospital, Zeytinburnu, 34010, Istanbul, Turkey.
| | - Yağmur Çetin-Taş
- Koç University Research Center for Translational Medicine, Istanbul, Turkey
| | - Emine Şekerdağ
- Koç University Research Center for Translational Medicine, Istanbul, Turkey
| | - Ali B Kızılırmak
- Koç University Research Center for Translational Medicine, Istanbul, Turkey
| | - Ali Taş
- Koç University Research Center for Translational Medicine, Istanbul, Turkey
| | - Erdost Yıldız
- Koç University Research Center for Translational Medicine, Istanbul, Turkey
| | - Hale Yapıcı-Eser
- Koç University Research Center for Translational Medicine, Istanbul, Turkey
- Department of Psychiatry, School of Medicine, Koç University, Istanbul, Turkey
| | - Serçin Karahüseyinoğlu
- Koç University Research Center for Translational Medicine, Istanbul, Turkey
- Department of Histology and Embryology, School of Medicine, Koç University, Istanbul, Turkey
| | - Müjdat Zeybel
- Koç University Research Center for Translational Medicine, Istanbul, Turkey
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust & University of Nottingham, Nottingham, UK
- Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Yasemin Gürsoy-Özdemir
- Koç University Research Center for Translational Medicine, Istanbul, Turkey
- Department of Neurology, School of Medicine, Koç University, Istanbul, Turkey
| |
Collapse
|
461
|
Hoyles L, Pontifex MG, Rodriguez-Ramiro I, Anis-Alavi MA, Jelane KS, Snelling T, Solito E, Fonseca S, Carvalho AL, Carding SR, Müller M, Glen RC, Vauzour D, McArthur S. Regulation of blood-brain barrier integrity by microbiome-associated methylamines and cognition by trimethylamine N-oxide. MICROBIOME 2021; 9:235. [PMID: 34836554 PMCID: PMC8626999 DOI: 10.1186/s40168-021-01181-z] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 10/18/2021] [Indexed: 05/09/2023]
Abstract
BACKGROUND Communication between the gut microbiota and the brain is primarily mediated via soluble microbe-derived metabolites, but the details of this pathway remain poorly defined. Methylamines produced by microbial metabolism of dietary choline and L-carnitine have received attention due to their proposed association with vascular disease, but their effects upon the cerebrovascular circulation have hitherto not been studied. RESULTS Here, we use an integrated in vitro/in vivo approach to show that physiologically relevant concentrations of the dietary methylamine trimethylamine N-oxide (TMAO) enhanced blood-brain barrier (BBB) integrity and protected it from inflammatory insult, acting through the tight junction regulator annexin A1. In contrast, the TMAO precursor trimethylamine (TMA) impaired BBB function and disrupted tight junction integrity. Moreover, we show that long-term exposure to TMAO protects murine cognitive function from inflammatory challenge, acting to limit astrocyte and microglial reactivity in a brain region-specific manner. CONCLUSION Our findings demonstrate the mechanisms through which microbiome-associated methylamines directly interact with the mammalian BBB, with consequences for cerebrovascular and cognitive function. Video abstract.
Collapse
Affiliation(s)
- Lesley Hoyles
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK.
| | | | - Ildefonso Rodriguez-Ramiro
- Norwich Medical School, University of East Anglia, Norwich, UK
- Metabolic Syndrome Group, Madrid Institute for Advanced Studies (IMDEA) in Food, E28049, Madrid, Spain
| | - M Areeb Anis-Alavi
- Institute of Dentistry, Faculty of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Khadija S Jelane
- Institute of Dentistry, Faculty of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Tom Snelling
- Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Egle Solito
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Queen Mary University of London, London, UK
- Dipartimento di Medicina molecolare e Biotecnologie mediche, Federico II University, Naples, Italy
| | - Sonia Fonseca
- The Gut Microbes and Health Research Programme, The Quadram Institute, Norwich Research Park, Norwich, UK
| | - Ana L Carvalho
- The Gut Microbes and Health Research Programme, The Quadram Institute, Norwich Research Park, Norwich, UK
| | - Simon R Carding
- Norwich Medical School, University of East Anglia, Norwich, UK
- The Gut Microbes and Health Research Programme, The Quadram Institute, Norwich Research Park, Norwich, UK
| | - Michael Müller
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Robert C Glen
- Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - David Vauzour
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Simon McArthur
- Institute of Dentistry, Faculty of Medicine & Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
462
|
Semyachkina-Glushkovskaya O, Penzel T, Blokhina I, Khorovodov A, Fedosov I, Yu T, Karandin G, Evsukova A, Elovenko D, Adushkina V, Shirokov A, Dubrovskii A, Terskov A, Navolokin N, Tzoy M, Ageev V, Agranovich I, Telnova V, Tsven A, Kurths J. Night Photostimulation of Clearance of Beta-Amyloid from Mouse Brain: New Strategies in Preventing Alzheimer's Disease. Cells 2021; 10:3289. [PMID: 34943796 PMCID: PMC8699220 DOI: 10.3390/cells10123289] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/11/2022] Open
Abstract
The deposition of amyloid-β (Aβ) in the brain is a risk factor for Alzheimer's disease (AD). Therefore, new strategies for the stimulation of Aβ clearance from the brain can be useful in preventing AD. Transcranial photostimulation (PS) is considered a promising method for AD therapy. In our previous studies, we clearly demonstrated the PS-mediated stimulation of lymphatic clearing functions, including Aβ removal from the brain. There is increasing evidence that sleep plays an important role in Aβ clearance. Here, we tested our hypothesis that PS at night can stimulate Aβ clearance from the brain more effectively than PS during the day. Our results on healthy mice show that Aβ clearance from the brain occurs faster at night than during wakefulness. The PS course at night improves memory and reduces Aβ accumulation in the brain of AD mice more effectively than the PS course during the day. Our results suggest that night PS is a more promising candidate as an effective method in preventing AD than daytime PS. These data are an important informative platform for the development of new noninvasive and nonpharmacological technologies for AD therapy as well as for preventing Aβ accumulation in the brain of people with disorder of Aβ metabolism, sleep deficit, elderly age, and jet lag.
Collapse
Affiliation(s)
- Oxana Semyachkina-Glushkovskaya
- Institute of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany;
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Thomas Penzel
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
- Sleep Medicine Center, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Inna Blokhina
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Alexander Khorovodov
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Ivan Fedosov
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Tingting Yu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China;
- Collaborative Innovation Center for Biomedical Engineering, MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Georgy Karandin
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Arina Evsukova
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Dariya Elovenko
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Viktoria Adushkina
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Alexander Shirokov
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
- Saratov Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), Institute of Biochemistry and Physiology of Plants and Microorganisms, Prospekt Entuziastov 13, 410049 Saratov, Russia
| | - Alexander Dubrovskii
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Andrey Terskov
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Nikita Navolokin
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
- Department of Pathological Anatomy, Saratov Medical State University, Kazachaya 112, 410012 Saratov, Russia
| | - Maria Tzoy
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Vasily Ageev
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Ilana Agranovich
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Valeria Telnova
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Anna Tsven
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
| | - Jürgen Kurths
- Institute of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany;
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (T.P.); (I.B.); (A.K.); (I.F.); (G.K.); (A.E.); (D.E.); (V.A.); (A.S.); (A.D.); (A.T.); (N.N.); (M.T.); (V.A.); (I.A.); (V.T.); (A.T.)
- Department of Complexity Science, Potsdam Institute for Climate Impact Research, Telegrafenberg A31, 14473 Potsdam, Germany
| |
Collapse
|
463
|
Hwang DJ, Choi DH, Kwon KC, Kim EH, Kim TK, Koo JH, Cho JY. Exercise Reverses Amyloid Beta-Peptide-mediated Cognitive Deficits in Alzheimer's Disease Mice Expressing Mutant Presenilin-2. Med Sci Sports Exerc 2021; 54:551-565. [PMID: 34816813 DOI: 10.1249/mss.0000000000002834] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PURPOSE The molecular mechanisms by which physical exercise produces beneficial effects on pathological features and behavioral symptoms of Alzheimer's disease (AD) are not well understood. Herein, we examined whether regular moderate exercise could improve cognitive function and produce transcriptomic responses in the brain. METHODS Four groups of mice were studied: non-transgenic control (Non-Tg), mice expressing the human presenilin-2 wild type (Tg-PS2w), mice expressing the human presenilin-2 with the N141I mutation (Tg-PS2m), and Tg-PS2m that were subjected to treadmill exercise (TE) at a speed of 10 m/min for 50 min/day, 5 days/week, for 6 weeks (Tg-PS2m/Ex). RESULTS Tg-PS2m/Ex mice exhibited increased preference in exploring a novel object than Tg-PS2m in the novel object recognition test (NORT), whereas differences observed in the water maze test and passive avoidance test were not significant. Western blot and histological analyses using amyloid oligomer (A11) and Aβ (6E10) antibody indicated that amyloid oligomer-reactive bands and plaque deposition in the hippocampus were reduced, though not significantly, after TE. Transcriptomic (RNA-sequencing) analysis and subsequent protein analysis revealed that the cell cycle regulatory gene, Cdc28 protein kinase regulatory subunit 2 (Cks2), was decreased, and the cell cycle- and apoptotic cell death-related factors, including cyclin D1, proliferating cell nuclear antigen, and cleaved caspase-3 were increased in the hippocampus of Tg-PS2m, whereas TE reversed their altered expression. CONCLUSION These results support the hypothesis that the pathological features and behavioral symptoms of AD caused by accumulation of amyloid beta-peptide in hippocampus, causing aberrant cell cycle re-entry and apoptosis, can be reversed by regular exercise.
Collapse
Affiliation(s)
- Dong-Joo Hwang
- Exercise Biochemistry Laboratory, Korea National Sport University, Seoul, Republic of Korea Department of Physical Education, Dongguk University, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
464
|
Sierra C, De Toma I, Cascio LL, Vegas E, Dierssen M. Social Factors Influence Behavior in the Novel Object Recognition Task in a Mouse Model of Down Syndrome. Front Behav Neurosci 2021; 15:772734. [PMID: 34803627 PMCID: PMC8602686 DOI: 10.3389/fnbeh.2021.772734] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/05/2021] [Indexed: 11/24/2022] Open
Abstract
The use of mouse models has revolutionized the field of Down syndrome (DS), increasing our knowledge about neuropathology and helping to propose new therapies for cognitive impairment. However, concerns about the reproducibility of results in mice and their translatability to humans have become a major issue, and controlling for moderators of behavior is essential. Social and environmental factors, the experience of the researcher, and the sex and strain of the animals can all have effects on behavior, and their impact on DS mouse models has not been explored. Here we analyzed the influence of a number of social and environmental factors, usually not taken into consideration, on the behavior of male and female wild-type and trisomic mice (the Ts65Dn model) in one of the most used tests for proving drug effects on memory, the novel object recognition (NOR) test. Using principal component analysis and correlation matrices, we show that the ratio of trisomic mice in the cage, the experience of the experimenter, and the timing of the test have a differential impact on male and female and on wild-type and trisomic behavior. We conclude that although the NOR test is quite robust and less susceptible to environmental influences than expected, to obtain useful results, the phenotype expression must be contrasted against the influences of social and environmental factors.
Collapse
Affiliation(s)
- Cesar Sierra
- Center for Genomic Regulation, The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Ilario De Toma
- Neurosciences Research Program, Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Lorenzo Lo Cascio
- Center for Genomic Regulation, The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Esteban Vegas
- Department of Genetics, Microbiology and Statistics, Section of Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Mara Dierssen
- Center for Genomic Regulation, The Barcelona Institute for Science and Technology, Barcelona, Spain
- Department of Experimental and Health Sciences, University Pompeu Fabra, Barcelona, Spain
- Biomedical Research Networking Center for Rare Diseases (CIBERER), Barcelona, Spain
| |
Collapse
|
465
|
Verma V, Kumar MJV, Sharma K, Rajaram S, Muddashetty R, Manjithaya R, Behnisch T, Clement JP. Pharmacological intervention in young adolescents rescues synaptic physiology and behavioural deficits in Syngap1 +/- mice. Exp Brain Res 2021; 240:289-309. [PMID: 34739555 DOI: 10.1007/s00221-021-06254-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 10/21/2021] [Indexed: 01/04/2023]
Abstract
Haploinsufficiency in SYNGAP1 is implicated in intellectual disability (ID) and autism spectrum disorder (ASD) and affects the maturation of dendritic spines. The abnormal spine development has been suggested to cause a disbalance of excitatory and inhibitory (E/I) neurotransmission at distinct developmental periods. In addition, E/I imbalances in Syngap1+/- mice might be due to abnormalities in K+-Cl- co-transporter function (NKCC1, KCC2), in a maner similar to the murine models of Fragile-X and Rett syndromes. To study whether an altered intracellular chloride ion concentration represents an underlying mechanism of modified function of GABAergic synapses in Dentate Gyrus Granule Cells of Syngap1+/- recordings were performed at different developmental stages of the mice. We observed depolarised neurons at P14-15 as illustrated by decreased Cl- reversal potential in Syngap1+/- mice. The KCC2 expression was decreased compared to Wild-type (WT) mice at P14-15. The GSK-3β inhibitor, 6-bromoindirubin-3'-oxime (6BIO) that crosses the blood-brain barrier, was tested to restore the function of GABAergic synapses. We discovered that the intraperitoneal administration of 6BIO during the critical period or young adolescents [P30 to P80 (4-week to 10-week)] normalised an altered E/I balance, the deficits of synaptic plasticity, and behavioural performance like social novelty, anxiety, and memory of the Syngap1+/- mice. In summary, altered GABAergic function in Syngap1+/- mice is due to reduced KCC2 expression leading to an increase in the intracellular chloride concentration that can be counteracted by the 6BIO, which restored cognitive, emotional, and social symptoms by pharmacological intervention, particularly in adulthood.
Collapse
Affiliation(s)
- Vijaya Verma
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - M J Vijay Kumar
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - Kavita Sharma
- International Centre for Material Sciences, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - Sridhar Rajaram
- International Centre for Material Sciences, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - Ravi Muddashetty
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, 560065, India
| | - Ravi Manjithaya
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India.,Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - Thomas Behnisch
- Institutes of Brain Sciences, Fudan University, Shanghai, 200032, China
| | - James P Clement
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India.
| |
Collapse
|
466
|
Lambuk L, Iezhitsa I, Agarwal R, Agarwal P, Peresypkina A, Pobeda A, Ismail NM. Magnesium acetyltaurate prevents retinal damage and visual impairment in rats through suppression of NMDA-induced upregulation of NF-κB, p53 and AP-1 (c-Jun/c-Fos). Neural Regen Res 2021; 16:2330-2344. [PMID: 33818520 PMCID: PMC8354133 DOI: 10.4103/1673-5374.310691] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/01/2020] [Accepted: 12/02/2020] [Indexed: 12/22/2022] Open
Abstract
Magnesium acetyltaurate (MgAT) has been shown to have a protective effect against N-methyl-D-aspartate (NMDA)-induced retinal cell apoptosis. The current study investigated the involvement of nuclear factor kappa-B (NF-κB), p53 and AP-1 family members (c-Jun/c-Fos) in neuroprotection by MgAT against NMDA-induced retinal damage. In this study, Sprague-Dawley rats were randomized to undergo intravitreal injection of vehicle, NMDA or MgAT as pre-treatment to NMDA. Seven days after injections, retinal ganglion cells survival was detected using retrograde labelling with fluorogold and BRN3A immunostaining. Functional outcome of retinal damage was assessed using electroretinography, and the mechanisms underlying antiapoptotic effect of MgAT were investigated through assessment of retinal gene expression of NF-κB, p53 and AP-1 family members (c-Jun/c-Fos) using reverse transcription-polymerase chain reaction. Retinal phospho-NF-κB, phospho-p53 and AP-1 levels were evaluated using western blot assay. Rat visual functions were evaluated using visual object recognition tests. Both retrograde labelling and BRN3A immunostaining revealed a significant increase in the number of retinal ganglion cells in rats receiving intravitreal injection of MgAT compared with the rats receiving intravitreal injection of NMDA. Electroretinography indicated that pre-treatment with MgAT partially preserved the functional activity of NMDA-exposed retinas. MgAT abolished NMDA-induced increase of retinal phospho-NF-κB, phospho-p53 and AP-1 expression and suppressed NMDA-induced transcriptional activity of NF-κB, p53 and AP-1 family members (c-Jun/c-Fos). Visual object recognition tests showed that MgAT reduced difficulties in recognizing the visual cues (i.e. objects with different shapes) after NMDA exposure, suggesting that visual functions of rats were relatively preserved by pre-treatment with MgAT. In conclusion, pre-treatment with MgAT prevents NMDA induced retinal injury by inhibiting NMDA-induced neuronal apoptosis via downregulation of transcriptional activity of NF-κB, p53 and AP-1-mediated c-Jun/c-Fos. The experiments were approved by the Animal Ethics Committee of Universiti Teknologi MARA (UiTM), Malaysia, UiTM CARE No 118/2015 on December 4, 2015 and UiTM CARE No 220/7/2017 on December 8, 2017 and Ethics Committee of Belgorod State National Research University, Russia, No 02/20 on January 10, 2020.
Collapse
Affiliation(s)
- Lidawani Lambuk
- Center for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Selangor, Malaysia
| | - Igor Iezhitsa
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Renu Agarwal
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Puneet Agarwal
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Anna Peresypkina
- Department of Pharmacology and Clinical Pharmacology, Institute of Medicine, Belgorod State National Research University, Belgorod, Russia
| | - Anna Pobeda
- Department of Pharmacology and Clinical Pharmacology, Institute of Medicine, Belgorod State National Research University, Belgorod, Russia
| | - Nafeeza Mohd Ismail
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
467
|
Schizophrenia-associated LRRTM1 regulates cognitive behavior through controlling synaptic function in the mediodorsal thalamus. Mol Psychiatry 2021; 26:6912-6925. [PMID: 33981006 DOI: 10.1038/s41380-021-01146-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 01/08/2023]
Abstract
Reduced activity of the mediodorsal thalamus (MD) and abnormal functional connectivity of the MD with the prefrontal cortex (PFC) cause cognitive deficits in schizophrenia. However, the molecular basis of MD hypofunction in schizophrenia is not known. Here, we identified leucine-rich-repeat transmembrane neuronal protein 1 (LRRTM1), a postsynaptic cell-adhesion molecule, as a key regulator of excitatory synaptic function and excitation-inhibition balance in the MD. LRRTM1 is strongly associated with schizophrenia and is highly expressed in the thalamus. Conditional deletion of Lrrtm1 in the MD in adult mice reduced excitatory synaptic function and caused a parallel reduction in the afferent synaptic activity of the PFC, which was reversed by the reintroduction of LRRTM1 in the MD. Our results indicate that chronic reduction of synaptic strength in the MD by targeted deletion of Lrrtm1 functionally disengages the MD from the PFC and may account for cognitive, social, and sensorimotor gating deficits, reminiscent of schizophrenia.
Collapse
|
468
|
Huang H, Jiang N, Zhang YW, Lv JW, Wang HX, Lu C, Liu XM, Lu GH. Gastrodia elata blume ameliorates circadian rhythm disorder-induced mice memory impairment. LIFE SCIENCES IN SPACE RESEARCH 2021; 31:51-58. [PMID: 34689950 DOI: 10.1016/j.lssr.2021.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/30/2021] [Accepted: 07/22/2021] [Indexed: 06/13/2023]
Abstract
Circadian rhythm disorder (CRD) in space flight can lead to memory impairment, performance decrements and adverse health outcomes, the main manifestations of which are circadian desynchronization, sleep loss and insomnia. Sleep deprivation (SD) provide the means to evaluate these effects and the risks associated with CRD on ground. Gastrodia elata Blume (GEB) has beneficial effects on the treatment of sleep disturbances and memory loss. Fresh GEB (FG), an unprocessed raw tuber of GEB, has been used as functional health food in Asian countries for a long time. However, the research report of FG to ameliorate memory impairment caused by insomnia or lack of sleep is meager. In this study, ICR male mice were sleep-deprived continuously and water extract of FG (WFG) was orally administrated (3 and 9 g/kg/d, i.g) during the SD process lasted for 25 days, except control and model groups gavage administration with water, positive control group with modafinil (MOD, 0.1 g/kg/d, i.g). We studied the effect of WFG on CRD-induced learning and memory impairment using a set of behavioral analyses including the object location recognition test (OLRT), novel object recognition test (NORT), and the passive avoidance test (PAT). In addition, oxidative stress parameters were assessed by measuring the malondialdehyde (MDA) and superoxide dismutase (SOD) reactivity in serum and hippocampus. Our results revealed that SD decreased discrimination index (DI) in OLRT and NORT, with shorter latency into the dark chamber in PAT. Both WFG and MOD treatment can reverse these changes (P < 0.05). We concluded that WFG treatment improve CRD-induced learning and memory impairment and oxidative stress damage which makes FG a promising candidate as herbal health product of memory decline in CRD.
Collapse
Affiliation(s)
- Hong Huang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Ning Jiang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Yi Wen Zhang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Jing Wei Lv
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Hai Xia Wang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Cong Lu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Xin Min Liu
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.
| | - Guang Hua Lu
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
469
|
Wu R, Liu J, Seaman R, Johnson B, Zhang Y, Li JX. The selective TAAR1 partial agonist RO5263397 promoted novelty recognition memory in mice. Psychopharmacology (Berl) 2021; 238:3221-3228. [PMID: 34291306 PMCID: PMC8605990 DOI: 10.1007/s00213-021-05937-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/09/2021] [Indexed: 11/28/2022]
Abstract
RATIONALE Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor that has a particular role in regulating dopaminergic, serotonergic, and glutamatergic transmission. TAAR1 agonists have shown pro-cognitive activities. However, it remains largely unknown of the effects of TAAR1 agonists on memory performance. OBJECTIVES Here, by using the mice novel object recognition (NOR) test, we examined the effects of the selective TAAR1 partial agonist RO5263397 on recognition memory. RESULTS We found that RO5263397 significantly enhanced the retrieval of short-term memory (STM; 20 min after training) both in male and female mice. RO5263397 promoted the retrieval of STM in the wild-type (WT) littermates but not TAAR1-KO mice, indicating that the effects of RO5263397 were dependent on TAAR1. Interestingly, compared to their WT litters, TAAR1-KO mice showed similar levels of STM, suggesting that genetic deletion of taar1 gene did not affect the STM retrieval. Furthermore, RO5263397 also promoted the retrieval of long-term NOR memory (24 h after training). CONCLUSIONS These results indicate that TAAR1 activation promotes NOR memory retrieval. Consistent with previous studies, our finding further suggests that TAAR1 agonists have pro-cognitive properties.
Collapse
Affiliation(s)
- Ruyan Wu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA.,Medical college of Yangzhou University, Yangzhou, China
| | - Jianfeng Liu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA
| | - Robert Seaman
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA
| | - Bernard Johnson
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, NC, USA
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University At Buffalo, The State University of New York, 955 Main Street, Buffalo, NY, 14214, USA.
| |
Collapse
|
470
|
Lorena FB, do Nascimento BPP, Camargo ELRA, Bernardi MM, Fukushima AR, do N Panizza J, de B Nogueira P, Brandão MES, Ribeiro MO. Long-term obesity is associated with depression and neuroinflammation. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2021; 65:537-548. [PMID: 34714995 PMCID: PMC10528574 DOI: 10.20945/2359-3997000000400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 06/18/2021] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Obesity is characterized by a state of chronic, low-intensity systemic inflammation frequently associated with insulin resistance and dyslipidemia. METHODS Given that chronic inflammation has been implicated in the pathogenesis of mood disorders, we investigated if chronic obesity that was initiated early in life - lasting through adulthood - could be more harmful to memory impairment and mood fluctuations such as depression. RESULTS Here we show that pre-pubertal male rats (30 days old) treated with a high-fat diet (40%) for 8-months gained ~50% more weight when compared to controls, exhibited depression and anxiety-like behaviors but no memory impairment. The prefrontal cortex of the obese rats exhibited an increase in the expression of genes related to inflammatory response, such as NFKb, MMP9, CCl2, PPARb, and PPARg. There were no alterations in genes known to be related to depression. CONCLUSION Long-lasting obesity with onset in prepuberal age led to depression and neuroinflammation but not to memory impairment.
Collapse
Affiliation(s)
- Fernanda B Lorena
- Programa de Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde, Universidade Presbiteriana Mackenzie, São Paulo, SP, Brasil
- Medicina Translacional, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - Bruna P P do Nascimento
- Programa de Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde, Universidade Presbiteriana Mackenzie, São Paulo, SP, Brasil
- Medicina Translacional, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - Esther L R A Camargo
- Programa de Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde, Universidade Presbiteriana Mackenzie, São Paulo, SP, Brasil
- Departamento de Pesquisa e Extensão, Faculdade de Ciências da Saúde IGESP, São Paulo, SP, Brasil
| | - Maria M Bernardi
- Instituto de Ciências da Saúde, Universidade Paulista, São Paulo, SP, Brasil
| | - André R Fukushima
- Departamento de Pesquisa e Extensão, Faculdade de Ciências da Saúde IGESP, São Paulo, SP, Brasil
| | - Julia do N Panizza
- Programa de Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde, Universidade Presbiteriana Mackenzie, São Paulo, SP, Brasil
| | - Paula de B Nogueira
- Programa de Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde, Universidade Presbiteriana Mackenzie, São Paulo, SP, Brasil
| | - Marllos E S Brandão
- Programa de Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde, Universidade Presbiteriana Mackenzie, São Paulo, SP, Brasil
- Medicina Translacional, Universidade Federal de São Paulo, São Paulo, SP, Brasil
- Departamento de Pesquisa e Extensão, Faculdade de Ciências da Saúde IGESP, São Paulo, SP, Brasil
| | - Miriam O Ribeiro
- Programa de Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde, Universidade Presbiteriana Mackenzie, São Paulo, SP, Brasil,
- Medicina Translacional, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
471
|
Cao J, Yao D, Li R, Guo X, Hao J, Xie M, Li J, Pan D, Luo X, Yu Z, Wang M, Wang W. Digoxin Ameliorates Glymphatic Transport and Cognitive Impairment in a Mouse Model of Chronic Cerebral Hypoperfusion. Neurosci Bull 2021; 38:181-199. [PMID: 34704235 PMCID: PMC8821764 DOI: 10.1007/s12264-021-00772-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 07/04/2021] [Indexed: 02/03/2023] Open
Abstract
The glymphatic system plays a pivotal role in maintaining cerebral homeostasis. Chronic cerebral hypoperfusion, arising from small vessel disease or carotid stenosis, results in cerebrometabolic disturbances ultimately manifesting in white matter injury and cognitive dysfunction. However, whether the glymphatic system serves as a potential therapeutic target for white matter injury and cognitive decline during hypoperfusion remains unknown. Here, we established a mouse model of chronic cerebral hypoperfusion via bilateral common carotid artery stenosis. We found that the hypoperfusion model was associated with significant white matter injury and initial cognitive impairment in conjunction with impaired glymphatic system function. The glymphatic dysfunction was associated with altered cerebral perfusion and loss of aquaporin 4 polarization. Treatment of digoxin rescued changes in glymphatic transport, white matter structure, and cognitive function. Suppression of glymphatic functions by treatment with the AQP4 inhibitor TGN-020 abolished this protective effect of digoxin from hypoperfusion injury. Our research yields new insight into the relationship between hemodynamics, glymphatic transport, white matter injury, and cognitive changes after chronic cerebral hypoperfusion.
Collapse
Affiliation(s)
- Jie Cao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Di Yao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Rong Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China ,Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Xuequn Guo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China ,Department of Respiratory Medicine, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, 362000 China
| | - Jiahuan Hao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Jia Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Dengji Pan
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Xiang Luo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Zhiyuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Minghuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China ,Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| |
Collapse
|
472
|
Pimpinella D, Mastrorilli V, Giorgi C, Coemans S, Lecca S, Lalive AL, Ostermann H, Fuchs EC, Monyer H, Mele A, Cherubini E, Griguoli M. Septal cholinergic input to CA2 hippocampal region controls social novelty discrimination via nicotinic receptor-mediated disinhibition. eLife 2021; 10:65580. [PMID: 34696824 PMCID: PMC8547952 DOI: 10.7554/elife.65580] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 09/30/2021] [Indexed: 12/03/2022] Open
Abstract
Acetylcholine (ACh), released in the hippocampus from fibers originating in the medial septum/diagonal band of Broca (MSDB) complex, is crucial for learning and memory. The CA2 region of the hippocampus has received increasing attention in the context of social memory. However, the contribution of ACh to this process remains unclear. Here, we show that in mice, ACh controls social memory. Specifically, MSDB cholinergic neurons inhibition impairs social novelty discrimination, meaning the propensity of a mouse to interact with a novel rather than a familiar conspecific. This effect is mimicked by a selective antagonist of nicotinic AChRs delivered in CA2. Ex vivo recordings from hippocampal slices provide insight into the underlying mechanism, as activation of nAChRs by nicotine increases the excitatory drive to CA2 principal cells via disinhibition. In line with this observation, optogenetic activation of cholinergic neurons in MSDB increases the firing of CA2 principal cells in vivo. These results point to nAChRs as essential players in social novelty discrimination by controlling inhibition in the CA2 region.
Collapse
Affiliation(s)
- Domenico Pimpinella
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Valentina Mastrorilli
- Department of Biology and Biotechnology 'C. Darwin', Center for Research in Neurobiology 'D. Bovet', Sapienza University of Rome, Rome, Italy
| | - Corinna Giorgi
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Rome, Italy.,Institute of Molecular Biology and Pathology of the National Council of Research (IBPM-CNR), Roma, Italy
| | - Silke Coemans
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Salvatore Lecca
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Arnaud L Lalive
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Hannah Ostermann
- Department of Clinical Neurobiology of the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elke C Fuchs
- Department of Clinical Neurobiology of the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hannah Monyer
- Department of Clinical Neurobiology of the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andrea Mele
- Department of Biology and Biotechnology 'C. Darwin', Center for Research in Neurobiology 'D. Bovet', Sapienza University of Rome, Rome, Italy
| | - Enrico Cherubini
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Marilena Griguoli
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Rome, Italy.,Institute of Neuroscience of the National Research Council (IN-CNR), Pisa, Italy
| |
Collapse
|
473
|
Pauls E, Bayod S, Mateo L, Alcalde V, Juan-Blanco T, Sánchez-Soto M, Saido TC, Saito T, Berrenguer-Llergo A, Attolini CSO, Gay M, de Oliveira E, Duran-Frigola M, Aloy P. Identification and drug-induced reversion of molecular signatures of Alzheimer's disease onset and progression in App NL-G-F, App NL-F, and 3xTg-AD mouse models. Genome Med 2021; 13:168. [PMID: 34702310 PMCID: PMC8547095 DOI: 10.1186/s13073-021-00983-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 09/29/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND In spite of many years of research, our understanding of the molecular bases of Alzheimer's disease (AD) is still incomplete, and the medical treatments available mainly target the disease symptoms and are hardly effective. Indeed, the modulation of a single target (e.g., β-secretase) has proven to be insufficient to significantly alter the physiopathology of the disease, and we should therefore move from gene-centric to systemic therapeutic strategies, where AD-related changes are modulated globally. METHODS Here we present the complete characterization of three murine models of AD at different stages of the disease (i.e., onset, progression and advanced). We combined the cognitive assessment of these mice with histological analyses and full transcriptional and protein quantification profiling of the hippocampus. Additionally, we derived specific Aβ-related molecular AD signatures and looked for drugs able to globally revert them. RESULTS We found that AD models show accelerated aging and that factors specifically associated with Aβ pathology are involved. We discovered a few proteins whose abundance increases with AD progression, while the corresponding transcript levels remain stable, and showed that at least two of them (i.e., lfit3 and Syt11) co-localize with Aβ plaques in the brain. Finally, we found two NSAIDs (dexketoprofen and etodolac) and two anti-hypertensives (penbutolol and bendroflumethiazide) that overturn the cognitive impairment in AD mice while reducing Aβ plaques in the hippocampus and partially restoring the physiological levels of AD signature genes to wild-type levels. CONCLUSIONS The characterization of three AD mouse models at different disease stages provides an unprecedented view of AD pathology and how this differs from physiological aging. Moreover, our computational strategy to chemically revert AD signatures has shown that NSAID and anti-hypertensive drugs may still have an opportunity as anti-AD agents, challenging previous reports.
Collapse
Affiliation(s)
- Eduardo Pauls
- Joint IRB-BSC-CRG Programme in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Sergi Bayod
- Joint IRB-BSC-CRG Programme in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Lídia Mateo
- Joint IRB-BSC-CRG Programme in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Víctor Alcalde
- Joint IRB-BSC-CRG Programme in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Teresa Juan-Blanco
- Joint IRB-BSC-CRG Programme in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Marta Sánchez-Soto
- Joint IRB-BSC-CRG Programme in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Antoni Berrenguer-Llergo
- Biostatistics and Bioinformatics Unit, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Camille Stephan-Otto Attolini
- Biostatistics and Bioinformatics Unit, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Marina Gay
- Proteomics Unit, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | | | - Miquel Duran-Frigola
- Joint IRB-BSC-CRG Programme in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Patrick Aloy
- Joint IRB-BSC-CRG Programme in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain.
| |
Collapse
|
474
|
Hayward GC, Caceres D, Copeland EN, Baranowski BJ, Mohammad A, Whitley KC, Fajardo VA, MacPherson REK. Characterization of Alzheimer's disease-like neuropathology in Duchenne's muscular dystrophy using the DBA/2J mdx mouse model. FEBS Open Bio 2021; 12:154-162. [PMID: 34668666 PMCID: PMC8727939 DOI: 10.1002/2211-5463.13317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 09/28/2021] [Accepted: 10/19/2021] [Indexed: 11/26/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle wasting disorder caused by a mutation in the dystrophin gene. In addition to muscle pathology, some patients with DMD will exhibit cognitive impairments with severity being linked to age and type of genetic mutation. Likewise, some studies have shown that mdx mice display impairments in spatial memory compared with wild‐type (WT) controls, while others have not observed any such effect. Most studies have utilized the traditional C57BL/10 (C57) mdx mouse, which exhibits a mild disease phenotype. Recently, the DBA/2J (D2) mdx mouse has emerged as a more severe and perhaps clinically relevant DMD model; however, studies examining cognitive function in these mice are limited. Thus, in this study we examined cognitive function in age‐matched C57 and D2 mdx mice along with their respective WT controls. Our findings show that 8‐ to 12‐week‐old C57 mdx mice did not display any differences in exploration time when challenged with a novel object recognition test. Conversely, age‐matched D2 mdx mice spent less time exploring objects in total as a well as less time exploring the novel object, suggestive of impaired recognition memory. Biochemical analyses of the D2 mdx brain revealed higher soluble amyloid precursor protein β (APPβ) and APP in the prefrontal cortex of mdx mice compared with WT, and lower soluble APPα in the hippocampus, suggestive of a shift towards amyloidogenesis and a similar pathogenesis to Alzheimer's disease. Furthermore, our study demonstrates the utility of the D2 mdx model in studying cognitive impairment.
Collapse
Affiliation(s)
| | - Daniela Caceres
- Faculty of Medicine, University of del Rosario, Bogota, Colombia
| | - Emily N Copeland
- Department of Kinesiology, Brock University, St. Catharines, Canada
| | | | - Ahmad Mohammad
- Department of Health Sciences, Brock University, St. Catharines, Canada
| | | | - Val A Fajardo
- Department of Kinesiology, Brock University, St. Catharines, Canada
| | | |
Collapse
|
475
|
Miyata S, Kashio T, Tsuchiya K, Mitsui S. Motopsin deficiency imparts partial insensitivity to doxorubicin-induced hippocampal impairments in adult mice. Neurosci Lett 2021; 763:136181. [PMID: 34416345 DOI: 10.1016/j.neulet.2021.136181] [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: 04/04/2021] [Revised: 08/12/2021] [Accepted: 08/14/2021] [Indexed: 10/20/2022]
Abstract
Motopsin is a serine protease that plays a crucial role in synaptic functions. Loss of motopsin function causes severe intellectual disability in humans. In this study, we evaluated the role of motopsin in the neuropathological development of cognitive impairments following chemotherapy, also known as chemobrain. Motopsin knockout (KO) and wild-type (WT) mice were intravenously injected with doxorubicin (Dox) or saline four times every 8 days and were evaluated for open field, novel object recognition, and passive avoidance tests. Parvalbumin-positive neurons in the hippocampus were immunohistochemically analyzed. Dox administration significantly decreased the total distance in the open field test in both WT and motopsin KO mice without affecting the duration spent in the center square. A significant interaction between the genotype and drug treatment was detected in the recognition index (the rate to investigate a novel object) in the novel object recognition test, although Dox treatment did not affect the total investigation time. Additionally, Dox treatment significantly decreased the recognition index in WT mice, whereas it tended to increase the recognition index in motopsin KO mice. Dox treatment did not affect the latency to enter a dark compartment in either WT or motopsin KO mice in the passive avoidance test. Interestingly, Dox treatment increased the parvalbumin-positive neurons in the stratum oriens of the hippocampus CA1 region of only WT mice, not motopsin KO mice. Our data suggest that motopsin deficiency imparted partial insensitivity to Dox-induced hippocampal impairments. Alternatively, motopsin may contribute to the neuropathology of chemobrain.
Collapse
Affiliation(s)
- Shiori Miyata
- Department of Rehabilitation Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa, Maebashi, Gunma 371-8514, Japan
| | - Taiki Kashio
- Department of Rehabilitation Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa, Maebashi, Gunma 371-8514, Japan
| | - Kenji Tsuchiya
- Department of Rehabilitation Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa, Maebashi, Gunma 371-8514, Japan
| | - Shinichi Mitsui
- Department of Rehabilitation Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa, Maebashi, Gunma 371-8514, Japan.
| |
Collapse
|
476
|
Salem MA, Budzyńska B, Kowalczyk J, El Sayed NS, Mansour SM. Tadalafil and bergapten mitigate streptozotocin-induced sporadic Alzheimer's disease in mice via modulating neuroinflammation, PI3K/Akt, Wnt/β-catenin, AMPK/mTOR signaling pathways. Toxicol Appl Pharmacol 2021; 429:115697. [PMID: 34428446 DOI: 10.1016/j.taap.2021.115697] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 12/17/2022]
Abstract
Sporadic Alzheimer's disease (SAD) is a slowly progressive neurodegenerative disorder. This study aimed to investigate neuroprotective potential of tadalafil (TAD) and bergapten (BG) in SAD-induced cognitive impairment in mice. SAD was induced by single injection of streptozotocin (STZ; 3 mg/kg, ICV). STZ resulted in AD-like pathologies including Aβ deposition, tau aggregation, impaired insulin and Wnt/β-catenin signaling, as well as autophagic dysfunction and neuroinflammation. Administration of TAD or BG at doses of 20 and 25 mg/kg, respectively, for 21 consecutive days attenuated STZ-induced hippocampal insult, preserved neuronal integrity, and improved cognitive function in the Morris water maze and object recognition tests paralleled by reduction in Aβ expression by 79 and 89% and tau hyperphosphorylation by 60 and 61%, respectively. TAD and BG also enhanced protein expression of pAkt, pGSK-3β, beclin-1 and methylated protein phosphatase 2A (PP2A) and gene expression of cyclin D1, while raised BDNF immunoreactivity. Furthermore, TAD and BG boosted hippocampal levels of cGMP, PKG, Wnt3a, and AMPK and reduced expression of β-catenin and mTOR by 74% and 51%, respectively. TAD and BG also halted neuroinflammation by reducing IL-23 and IL-27 levels, as well as protein expression of NF-κB by 62% & 61%, respectively. In conclusion, this study offers novel insights on the neuroprotective effects of TAD or BG in the management of SAD as evidenced by improved cognitive function and histological architecture. This could be attributed to modulation of the crosstalk among PI3K/Akt/GSK-3β, PP2A, mTOR/autophagy, cGMP/PKG, and Wnt/β-catenin signaling cascades and mitigation of neuroinflammation.
Collapse
Affiliation(s)
- Mohamed A Salem
- Department of Pharmacology, Toxicology and Biochemistry, Faculty of Pharmacy, Future University in Egypt, Cairo, Egypt
| | - Barbara Budzyńska
- Independent Laboratory of Behavioral Studies, Medical University of Lublin, Lublin, Poland
| | - Joanna Kowalczyk
- Independent Laboratory of Behavioral Studies, Medical University of Lublin, Lublin, Poland; Chair and Department of Applied Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Nesrine S El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Suzan M Mansour
- Department of Pharmacology, Toxicology and Biochemistry, Faculty of Pharmacy, Future University in Egypt, Cairo, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| |
Collapse
|
477
|
Beaver JN, Gilman TL. Salt as a non-caloric behavioral modifier: A review of evidence from pre-clinical studies. Neurosci Biobehav Rev 2021; 135:104385. [PMID: 34634356 DOI: 10.1016/j.neubiorev.2021.10.007] [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: 08/04/2021] [Revised: 09/16/2021] [Accepted: 10/04/2021] [Indexed: 12/18/2022]
Abstract
Though excess salt intake is well-accepted as a dietary risk factor for cardiovascular diseases, relatively little has been explored about how it impacts behavior, despite the ubiquity of salt in modern diets. Given the challenges of manipulating salt intake in humans, non-human animals provide a more tractable means for evaluating behavioral sequelae of high salt. By describing what is known about the impact of elevated salt on behavior, this review highlights how underexplored salt's behavioral effects are. Increased salt consumption in adulthood does not affect spontaneous anxiety-related behaviors or locomotor activity, nor acquisition of maze or fear tasks, but does impede expression of spatial/navigational and fear memory. Nest building is reduced by heightened salt in adults, and stress responsivity is augmented. When excess salt exposure occurs during development, and/or to parents, offspring locomotion is increased, and both spatial memory expression and social investigation are attenuated. The largely consistent findings reviewed here indicate expanded study of salt's effects will likely uncover broader behavioral implications, particularly in the scarcely studied female sex.
Collapse
Affiliation(s)
- Jasmin N Beaver
- Department of Psychological Sciences & Brain Health Research Institute, Kent State University, Kent, OH, 44242, USA.
| | - T Lee Gilman
- Department of Psychological Sciences & Brain Health Research Institute, Kent State University, Kent, OH, 44242, USA.
| |
Collapse
|
478
|
Nanonaringenin and Vitamin E Ameliorate Some Behavioral, Biochemical, and Brain Tissue Alterations Induced by Nicotine in Rats. J Toxicol 2021; 2021:4411316. [PMID: 34608387 PMCID: PMC8487377 DOI: 10.1155/2021/4411316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 09/11/2021] [Indexed: 02/06/2023] Open
Abstract
Nicotine is the major alkaloid present in cigarettes that induces various biochemical and behavioral changes. Nanonaringenin (NNG) and vitamin E are antioxidants that are reported to mitigate serious impairments caused by some toxins and oxidants. Thus, we aimed to investigate the efficacy of NNG, vitamin E, and their combinations to ameliorate behavioral, biochemical, and histological alterations induced by nicotine in rats. Adult male albino rats were randomly grouped into six equal groups (10 rats/group): control, N (nicotine 1 mg/kg b.w./day S/C from 15th to 45th day, 5 days a week), NNG (25 mg/kg b.w./day orally for 45 days), N + NNG, N + E (nicotine + vitamin E 200 mg/kg b.w./day orally), and N + NNG + E (nicotine + NNG + vitamin E at the aforementioned doses). Behavioral tests were conducted on day 15 and 30 postnicotine injection, while memory tests, brain neurotransmitters, antioxidants, and histopathological examination were examined at day 30 only. As a result, nicotine impaired rats' activity (hypoactivity and hyperactivity) and memory, induced anxiolytic and anxiogenic effects on rats, and altered neurotransmitters (acetylcholinesterase, serotonin, and dopamine), and redox markers (MDA, H2O2, GSH, and catalase) levels in brain homogenates. Thickening and congestion of the meninges and degeneration of the cerebral neurons and glia cells were observed. Cosupplementation with NNG, vitamin E, and their combination with nicotine was beneficial in the alleviation of activity impairments and improved short memory and cognition defects and exploratory behaviors. Our results indicate the antioxidant potential of NNG and vitamin E by modulating redox markers and neurotransmitters in the brain. Thus, data suggest that the prophylactic use of NNG, vitamin E, and/or their combination for (45 days) may have a successful amelioration of the disrupted behavior and cognition and biochemical and histopathological alterations induced by nicotine.
Collapse
|
479
|
Zhang S, Xu Y, Zeng L, An X, Su D, Qu Y, Ma J, Tang X, Wang X, Yang J, Mishra C, Chandra SR, Ai J. Epigallocatechin-3-Gallate Allosterically Activates Protein Kinase C-α and Improves the Cognition of Estrogen Deficiency Mice. ACS Chem Neurosci 2021; 12:3672-3682. [PMID: 34505505 DOI: 10.1021/acschemneuro.1c00401] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Protein kinase C (PKC) isozymes play essential roles in biological processes, and activation of PKC is proposed to alleviate the symptoms of a variety of diseases. It would be of great significance to find effective pharmacological modulators of PKC isozymes that can be translated for clinical use. Here, using in vitro activity assay, we demonstrated that green tea extract (-)-epigallocatechin-3-gallate (EGCG) dose-dependently activated PKCα with a half effective concentration (EC50) of 0.49 μM. We also performed surface plasmon resonance analysis and found that EGCG binds PKCα with an equilibrium dissociation constant (KD) value of 4.11 × 10-6 mol/L. Further computational flexible docking analysis revealed that EGCG interacted with the catalytic C3-C4 domain of PKCα (PDB: 4RA4) through establishing polar hydrogen bonds with V420, T401, E387, and K368 of PKCα, and the benzene ring group of EGCG hydrophobically interacted with the hydrophobic pocket formed by L345, M470, I479, and V353 of PKCα. Interestingly, the PKCα-selective blocker Ro-32-0432 could compete with EGCG for the same substrate-binding pocket of PKCα. Moreover, we found that EGCG dose-dependently improved the spatial memory, object recognition ability, and hippocampal long-term potentiation of ovariectomized mice, which was offset by Ro-32-0432. Collectively, our findings reveal a novel PKCα agonist and open the way to a new perspective on PKCα pharmacology and the treatment of PKCα-related diseases, including cognitive impairment.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical University, Harbin, Heilongjiang Province 150086, China
| | - Yi Xu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical University, Harbin, Heilongjiang Province 150086, China
| | - Lu Zeng
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical University, Harbin, Heilongjiang Province 150086, China
| | - Xiaobin An
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical University, Harbin, Heilongjiang Province 150086, China
| | - Dan Su
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical University, Harbin, Heilongjiang Province 150086, China
| | - Yang Qu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical University, Harbin, Heilongjiang Province 150086, China
| | - Jing Ma
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical University, Harbin, Heilongjiang Province 150086, China
| | - Xin Tang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical University, Harbin, Heilongjiang Province 150086, China
| | - Xuqiao Wang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical University, Harbin, Heilongjiang Province 150086, China
| | - Junkai Yang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical University, Harbin, Heilongjiang Province 150086, China
| | - Chandan Mishra
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical University, Harbin, Heilongjiang Province 150086, China
| | - Shah Ram Chandra
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical University, Harbin, Heilongjiang Province 150086, China
| | - Jing Ai
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical University, Harbin, Heilongjiang Province 150086, China
| |
Collapse
|
480
|
Olson B, Zhu X, Norgard MA, Diba P, Levasseur PR, Buenafe AC, Huisman C, Burfeind KG, Michaelis KA, Kong G, Braun T, Marks DL. Chronic cerebral lipocalin 2 exposure elicits hippocampal neuronal dysfunction and cognitive impairment. Brain Behav Immun 2021; 97:102-118. [PMID: 34245812 PMCID: PMC8453133 DOI: 10.1016/j.bbi.2021.07.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/21/2021] [Accepted: 07/03/2021] [Indexed: 12/12/2022] Open
Abstract
Lipocalin 2 (LCN2) is a pleiotropic molecule that is induced in the central nervous system (CNS) in several acute and chronic pathologies. The acute induction of LCN2 evolved as a beneficial process, aimed at combating bacterial infection through the sequestration of iron from pathogens, while the role of LCN2 during chronic, non-infectious disease remains unclear, and recent studies suggest that LCN2 is neurotoxic. However, whether LCN2 is sufficient to induce behavioral and cognitive alterations remains unclear. In this paper, we sought to address the role of cerebral LCN2 on cognition in both acute and chronic settings. We demonstrate that LCN2 is robustly induced in the CNS during both acute and chronic inflammatory conditions, including LPS-based sepsis and cancer cachexia. In vivo, LPS challenge results in a global induction of LCN2 in the central nervous system, while cancer cachexia results in a distribution specific to the vasculature. Similar to these in vivo observations, in vitro modeling demonstrated that both glia and cerebral endothelium produce and secrete LCN2 when challenged with LPS, while only cerebral endothelium secrete LCN2 when challenged with cancer-conditioned medium. Chronic, but not short-term, cerebral LCN2 exposure resulted in reduced hippocampal neuron staining intensity, an increase in newborn neurons, microglial activation, and increased CNS immune cell infiltration, while gene set analyses suggested these effects were mediated through melanocortin-4 receptor independent mechanisms. RNA sequencing analyses of primary hippocampal neurons revealed a distinct transcriptome associated with prolonged LCN2 exposure, and ontology analysis was suggestive of altered neurite growth and abnormal spatial learning. Indeed, LCN2-treated hippocampal neurons display blunted neurite processes, and mice exposed to prolonged cerebral LCN2 levels experienced a reduction in spatial reference memory as indicated by Y-maze assessment. These findings implicate LCN2 as a pathologic mediator of cognitive decline in the setting of chronic disease.
Collapse
Affiliation(s)
- Brennan Olson
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR USA, Medical Scientist Training Program, Oregon Health & Science University, Portland, OR USA
| | - Xinxia Zhu
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR USA
| | - Mason A Norgard
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR USA
| | - Parham Diba
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR USA, Medical Scientist Training Program, Oregon Health & Science University, Portland, OR USA
| | - Peter R Levasseur
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR USA
| | - Abby C Buenafe
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR USA
| | - Christian Huisman
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR USA
| | - Kevin G Burfeind
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR USA, Medical Scientist Training Program, Oregon Health & Science University, Portland, OR USA
| | - Katherine A Michaelis
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR USA, Medical Scientist Training Program, Oregon Health & Science University, Portland, OR USA
| | - Garth Kong
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR USA
| | - Theodore Braun
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR USA
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR, USA; Brenden-Colson Center for Pancreatic Care, Oregon Health and & Science University Portland, OR, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
481
|
Ng RCL, Jian M, Ma OKF, Bunting M, Kwan JSC, Zhou GJ, Senthilkumar K, Iyaswamy A, Chan PK, Li M, Leung KMY, Kumar Durairajan SS, Lam KSL, Chu LW, Festenstein R, Chung SK, Chan KH. Chronic oral administration of adipoRon reverses cognitive impairments and ameliorates neuropathology in an Alzheimer's disease mouse model. Mol Psychiatry 2021; 26:5669-5689. [PMID: 32132650 DOI: 10.1038/s41380-020-0701-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 02/18/2020] [Accepted: 02/21/2020] [Indexed: 01/01/2023]
Abstract
Circulating adiponectin (APN) levels decrease with age and obesity. On the other hand, a reduction in APN levels is associated with neurodegeneration and neuroinflammation. We previously showed that aged adiponectin knockout (APN-/-) mice developed Alzheimer's like pathologies, cerebral insulin resistance, and cognitive impairments. More recently, we also demonstrated that APN deficiency increased Aβ-induced microglia activation and neuroinflammatory responses in 5xFAD mice. There is compelling evidence that deregulated insulin activities or cerebral insulin resistance contributes to neuroinflammation and Alzheimer's disease (AD) pathogenesis. Here, we demonstrated that APN levels were reduced in the brain of AD patients and 5xFAD mice. We crossbred 5xFAD mice with APN-/- mice to generate APN-deficient 5xFAD (5xFAD;APN-/-). APN deficiency in 5xFAD mice accelerated amyloid loading, increased cerebral amyloid angiopathy, and reduced insulin-signaling activities. Pharmacokinetics study demonstrated adipoRon (APN receptor agonist) was a blood-brain barrier penetrant. AdipoRon improved neuronal insulin-signaling activities and insulin sensitivity in vitro and in vivo. Chronic adipoRon treatment improved spatial memory functions and significantly rescued neuronal and synaptic loss in 5xFAD and 5xFAD;APN-/- mice. AdipoRon lowered plaque and Aβ levels in AD mice. AdipoRon also exerted anti-inflammatory effects by reducing microglial and astrocytes activation as well as suppressing cerebral cytokines levels. The microglial phagocytic activity toward Aβ was restored after adipoRon treatment. Our results indicated that adipoRon exerts multiple beneficial effects providing important therapeutic implications. We propose chronic adipoRon administration as a potential treatment for AD.
Collapse
Affiliation(s)
- Roy Chun-Laam Ng
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Neuroimmunology and Neuroinflammation Research Laboratory, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Min Jian
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Neuroimmunology and Neuroinflammation Research Laboratory, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Oscar Ka-Fai Ma
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Neuroimmunology and Neuroinflammation Research Laboratory, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Myriam Bunting
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Neuroimmunology and Neuroinflammation Research Laboratory, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jason Shing-Cheong Kwan
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Neuroimmunology and Neuroinflammation Research Laboratory, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Guang-Jie Zhou
- The Swire Institute of Marine Science and School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Krishnamoorthi Senthilkumar
- Mr & Mrs Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Ashok Iyaswamy
- Mr & Mrs Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Ping-Kei Chan
- Gene Control Mechanisms and Disease Group, Department of Brain Sciences, Faculty of Medicine, MRC London Institute of Medical Sciences, Imperial College London, London, W120NN, UK
| | - Min Li
- Mr & Mrs Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Kenneth Mei-Yee Leung
- The Swire Institute of Marine Science and School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Siva-Sundara Kumar Durairajan
- Mr & Mrs Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Division of Myobiology and Neurodegenerative Disease Research, Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur, India
| | - Karen Siu-Ling Lam
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Research Center of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Leung-Wing Chu
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Richard Festenstein
- Gene Control Mechanisms and Disease Group, Department of Brain Sciences, Faculty of Medicine, MRC London Institute of Medical Sciences, Imperial College London, London, W120NN, UK
| | - Sookja Kim Chung
- Research Center of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong, Pokfulam, Hong Kong, China
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Faculty of Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau
| | - Koon-Ho Chan
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
- Neuroimmunology and Neuroinflammation Research Laboratory, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
- Research Center of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong, Pokfulam, Hong Kong, China.
| |
Collapse
|
482
|
Wu D, Kumal JPP, Lu X, Li Y, Mao D, Tang X, Nie M, Liu X, Sun L, Liu B, Zhang Y. Traumatic Brain Injury Accelerates the Onset of Cognitive Dysfunction and Aggravates Alzheimer's-Like Pathology in the Hippocampus by Altering the Phenotype of Microglia in the APP/PS1 Mouse Model. Front Neurol 2021; 12:666430. [PMID: 34539542 PMCID: PMC8440856 DOI: 10.3389/fneur.2021.666430] [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: 02/10/2021] [Accepted: 07/15/2021] [Indexed: 11/18/2022] Open
Abstract
An increasing number of studies have suggested that traumatic brain injury (TBI) is associated with some neurodegenerative diseases, including Alzheimer's disease (AD). Various aspects of the mechanism of TBI-induced AD have been elucidated. However, there are also studies opposing the view that TBI is one of the causes of AD. In the present study, we demonstrated that TBI exacerbated the disruption of hippocampal-dependent learning and memory, worsened the reductions in neuronal cell density and synapse formation, and aggravated the deposition of Aβ plaques in the hippocampi of APP/PS1 mice. We also found that TBI rapidly activated microglia in the central nervous system (CNS) and that this effect lasted for at least for 3 weeks. Furthermore, TBI boosted Aβ-related microglia-mediated neuroinflammation in the hippocampi of APP/PS1 mice and the transformation of microglia toward the proinflammatory phenotype. Therefore, our experiments suggest that TBI accelerates the onset of cognitive dysfunction and Alzheimer-like pathology in the APP/PS1 mouse model, at least partly by altering microglial reactions and polarization.
Collapse
Affiliation(s)
- Di Wu
- Department of Neurology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jay Prakash P Kumal
- Department of Human Anatomy, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin, China
| | - Xiaodi Lu
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yixuan Li
- Department of Human Anatomy, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin, China
| | - Dongsheng Mao
- Department of Human Anatomy, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin, China
| | - Xudong Tang
- Department of Human Anatomy, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin, China
| | - Meitong Nie
- Department of Human Anatomy, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin, China
| | - Xin Liu
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang, China
| | - Liang Sun
- Department of Human Anatomy, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin, China
| | - Bin Liu
- Department of Neurology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yafang Zhang
- Department of Human Anatomy, Harbin Medical University, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin, China
| |
Collapse
|
483
|
Lissner LJ, Wartchow KM, Toniazzo AP, Gonçalves CA, Rodrigues L. Object recognition and Morris water maze to detect cognitive impairment from mild hippocampal damage in rats: A reflection based on the literature and experience. Pharmacol Biochem Behav 2021; 210:173273. [PMID: 34536480 DOI: 10.1016/j.pbb.2021.173273] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 10/20/2022]
Abstract
Object recognition (OR) and the Morris water maze (MWM) are classical tasks widely used to assess memory parameters and deficits in rodents. Learning processes in both tasks involve integrity of the hippocampus and associated regions, and prefrontal cortex connections. Here, we highlight the idea that these classical tests can be used to indicate memory deficits caused by models of disease that affect hippocampal function in rats, and identify some practical issues of OR and MWM, based on the literature and our experience. Additionally, we have shown that the performance of both tasks does not alter blood levels of corticosterone, considering exposure to a single task. Hence, taking into consideration the difficulties and care required during task execution, the infrastructure needed and the training of the experimenter, we suggest that OR and its variations offer minimal manageable stressful conditions, representing an effective and practical tool for hippocampal-related memory assessment of rats. Thus, OR may provide similar information to that of the MWM, despite controversy regarding hippocampus participation in OR and given due differences in the types of memory evaluated and researchers' objectives. We recommend the observation of some important precautions and details, also based on the literature and our own experience.
Collapse
Affiliation(s)
- Lílian Juliana Lissner
- Federal University of Rio Grande do Sul (UFRGS), Biochemistry Post-Graduate Program, Porto Alegre, Brazil
| | - Krista Minéia Wartchow
- Federal University of Rio Grande do Sul (UFRGS), Biochemistry Post-Graduate Program, Porto Alegre, Brazil
| | - Ana Paula Toniazzo
- Federal University of Rio Grande do Sul (UFRGS), Biochemistry Post-Graduate Program, Porto Alegre, Brazil
| | - Carlos-Alberto Gonçalves
- Federal University of Rio Grande do Sul (UFRGS), Biochemistry Post-Graduate Program, Porto Alegre, Brazil
| | - Leticia Rodrigues
- Federal University of Rio Grande do Sul (UFRGS), Biochemistry Post-Graduate Program, Porto Alegre, Brazil.
| |
Collapse
|
484
|
Qian C, Yang C, Lu M, Bao J, Shen H, Deng B, Li S, Li W, Zhang M, Cao C. Activating AhR alleviates cognitive deficits of Alzheimer's disease model mice by upregulating endogenous Aβ catabolic enzyme Neprilysin. Theranostics 2021; 11:8797-8812. [PMID: 34522212 PMCID: PMC8419060 DOI: 10.7150/thno.61601] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 08/02/2021] [Indexed: 02/07/2023] Open
Abstract
Rationale: Neprilysin (NEP) is a major endogenous catabolic enzyme of amyloid β (Aβ). Previous studies have suggested that increasing NEP expression in animal models of Alzheimer's disease had an ameliorative effect. However, the underlying signaling pathway that regulates NEP expression remains unclear. The aryl hydrocarbon receptor (AhR) is a ligand-activated cytoplasmic receptor and transcription factor. Recent studies have shown that AhR plays essential roles in the central nervous system (CNS), but its physiological and pathological roles in regulating NEP are not entirely known. Methods: Western blotting, immunofluorescence, quantitative RT-PCR and enzyme activity assay were used to verify the effects of AhR agonists on NEP in a cell model (N2a) and a mouse model (APP/PS1). Luciferase reporter assay and chromatin immunoprecipitation (ChIP) assay were conducted to investigate the roles of AhR in regulating NEP transcription. Object recognition test and the Morris water maze task were performed to assess the cognitive capacity of the mice. Results: Activating AhR by the endogenous ligand L-Kynurenine (L-KN) or FICZ, or by the exogenous ligand diosmin or indole-3-carbinol (I3C) significantly increases NEP expression and enzyme activity in N2a cells and APP/PS1 mice. We also found that AhR is a direct transcription factor of NEP. Diosmin treatment effectively ameliorated the cognitive disorder and memory deficit of APP/PS1 transgenic mice. By knocking down AhR or using a small molecular inhibitor targeting AhR or NEP, we found that diosmin enhanced Aβ degradation through activated AhR and increased NEP expression. Conclusions: These results indicate a novel pathway for regulating NEP expression in neurons and that AhR may be a potential therapeutic target for the treatment of Alzheimer's disease.
Collapse
|
485
|
Lai Z, Min J, Li J, Shan W, Yu W, Zuo Z. Surgery Trauma Severity but not Anesthesia Length Contributes to Postoperative Cognitive Dysfunction in Mice. J Alzheimers Dis 2021; 80:245-257. [PMID: 33523008 DOI: 10.3233/jad-201232] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Perioperative, modifiable factors contributing to perioperative neurocognitive disorders (PND) have not been clearly defined. OBJECTIVE To determine the contribution of anesthesia lengths and the degrees of surgical trauma to PND and neuroinflammation, a critical process for PND. METHODS Three-month-old C57BL/6J mice were subjected to 2 h or 6 h isoflurane anesthesia plus a 5 min or 15 min left common carotid artery exposure (surgery) in a factorial design (two factors: anesthesia with two levels and surgery with three levels). Their learning and memory were tested by Barnes maze and novel object recognition paradigms. Blood, spleen, and hippocampus were harvested for measuring interleukin (IL)-6 and IL-1β. Eighteen-month-old C57BL/6J mice (old mice) were subjected to 6 h isoflurane anesthesia or 2 h isoflurane anesthesia plus 15 min surgery and then had learning and memory tested. RESULTS Three-month-old mice with 15 min surgery (long surgery) under 2 h or 6 h anesthesia performed poorly in the learning and memory tests compared with controls. Anesthesia alone or anesthesia plus 5 min surgery did not affect mouse performance in these tests. Similarly, only mice with long surgery but not mice with other experimental conditions had increased IL-6 and IL-1β in the blood, spleen, and hippocampus and decreased spleen weights. Splenocytes were found in the hippocampus after surgery. Similarly, old mice with long surgery but not the mice with isoflurane anesthesia alone had poor performance in the Barnes maze and novel object recognition tests. CONCLUSION Surgical trauma, but not anesthesia, contributes to the development of PND and neuroinflammation. Splenocytes may modulate these processes.
Collapse
Affiliation(s)
- Zhongmeng Lai
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, USA.,Department of Anesthesiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jia Min
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, USA.,Department of Anesthesiology, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Jun Li
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, USA
| | - Weiran Shan
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, USA
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
486
|
Ames J, Yadavalli T, Suryawanshi R, Hopkins J, Agelidis A, Patil C, Fredericks B, Tseng H, Valyi-Nagy T, Shukla D. OPTN is a host intrinsic restriction factor against neuroinvasive HSV-1 infection. Nat Commun 2021; 12:5401. [PMID: 34518549 PMCID: PMC8437952 DOI: 10.1038/s41467-021-25642-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 08/11/2021] [Indexed: 11/09/2022] Open
Abstract
Fast-replicating neurotropic herpesviruses exemplified by herpes simplex virus-1 (HSV-1) naturally infect the central nervous system (CNS). However, most individuals intrinsically suppress the virus during a primary infection and preclude it from significantly damaging the CNS. Optineurin (OPTN) is a conserved autophagy receptor with little understanding of its role in neurotropic viral infections. We show that OPTN selectively targets HSV-1 tegument protein, VP16, and the fusion glycoprotein, gB, to degradation by autophagy. OPTN-deficient mice challenged with HSV-1 show significant cognitive decline and susceptibility to lethal CNS infection. OPTN deficiency unveils severe consequences for recruitment of adaptive immunity and suppression of neuronal necroptosis. Ocular HSV-1 infection is lethal without OPTN and is rescued using a necroptosis inhibitor. These results place OPTN at the crux of neuronal survival from potentially lethal CNS viral infections.
Collapse
Affiliation(s)
- Joshua Ames
- Department of Microbiology and Immunology, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA
| | - Tejabhiram Yadavalli
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA
| | - Rahul Suryawanshi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA
| | - James Hopkins
- Department of Microbiology and Immunology, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA
| | - Alexander Agelidis
- Department of Microbiology and Immunology, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA
| | - Chandrashekhar Patil
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA
| | - Brian Fredericks
- Department of Pathology, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA
| | - Henry Tseng
- Duke Eye Center and Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA
| | - Tibor Valyi-Nagy
- Department of Pathology, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA
| | - Deepak Shukla
- Department of Microbiology and Immunology, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA.
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA.
| |
Collapse
|
487
|
Promoting and Optimizing the Use of 3D-Printed Objects in Spontaneous Recognition Memory Tasks in Rodents: A Method for Improving Rigor and Reproducibility. eNeuro 2021; 8:ENEURO.0319-21.2021. [PMID: 34503967 PMCID: PMC8489023 DOI: 10.1523/eneuro.0319-21.2021] [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: 07/27/2021] [Revised: 08/28/2021] [Accepted: 09/01/2021] [Indexed: 11/21/2022] Open
Abstract
Spontaneous recognition memory tasks are widely used to assess cognitive function in rodents and have become commonplace in the characterization of rodent models of neurodegenerative, neuropsychiatric and neurodevelopmental disorders. Leveraging an animal’s innate preference for novelty, these tasks use object exploration to capture the what, where and when components of recognition memory. Choosing and optimizing objects is a key feature when designing recognition memory tasks. Although the range of objects used in these tasks varies extensively across studies, object features can bias exploration, influence task difficulty and alter brain circuit recruitment. Here, we discuss the advantages of using 3D-printed objects in rodent spontaneous recognition memory tasks. We provide strategies for optimizing their design and usage, and offer a repository of tested, open-source designs for use with commonly used rodent species. The easy accessibility, low-cost, renewability and flexibility of 3D-printed open-source designs make this approach an important step toward improving rigor and reproducibility in rodent spontaneous recognition memory tasks.
Collapse
|
488
|
Tao L, Liu Q, Zhang F, Fu Y, Zhu X, Weng X, Han H, Huang Y, Suo Y, Chen L, Gao X, Wei X. Microglia modulation with 1070-nm light attenuates Aβ burden and cognitive impairment in Alzheimer's disease mouse model. LIGHT, SCIENCE & APPLICATIONS 2021; 10:179. [PMID: 34493703 PMCID: PMC8423759 DOI: 10.1038/s41377-021-00617-3] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/29/2021] [Accepted: 08/17/2021] [Indexed: 05/05/2023]
Abstract
Photobiomodulation, by utilizing low-power light in the visible and near-infrared spectra to trigger biological responses in cells and tissues, has been considered as a possible therapeutic strategy for Alzheimer's disease (AD), while its specific mechanisms have remained elusive. Here, we demonstrate that cognitive and memory impairment in an AD mouse model can be ameliorated by 1070-nm light via reducing cerebral β-amyloid (Aβ) burden, the hallmark of AD. The glial cells, including microglia and astrocytes, play important roles in Aβ clearance. Our results show that 1070-nm light pulsed at 10 Hz triggers microglia rather than astrocyte responses in AD mice. The 1070-nm light-induced microglia responses with alteration in morphology and increased colocalization with Aβ are sufficient to reduce Aβ load in AD mice. Moreover, 1070-nm light pulsed at 10 Hz can reduce perivascular microglia and promote angiogenesis to further enhance Aβ clearance. Our study confirms the important roles of microglia and cerebral vessels in the use of 1070-nm light for the treatment of AD mice and provides a framework for developing a novel therapeutic approach for AD.
Collapse
Affiliation(s)
- Lechan Tao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Qi Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Fuli Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yuting Fu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xi Zhu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xiaofu Weng
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Hongbin Han
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China
- Key Lab of Magnetic Resonance Imaging Device and Technique, Beijing, 100191, China
| | - Yong Huang
- Zhejiang Brainhealth Medical Technology Co., Ltd, Hangzhou, 314400, China
| | - Yuanzhen Suo
- Biomedical Pioneering Innovation Center, Peking University, Beijing, 100871, China
- School of Life Sciences, Peking University, Beijing, 100871, China
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China.
- Tianqiao and Chrissy Chen Institute for Clinical Translational Research, Huashan Hospital, Shanghai, 200040, China.
| | - Xiaoling Gao
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xunbin Wei
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China.
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.
- Biomedical Engineering Department, Peking University, Beijing, 100081, China.
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
489
|
Carpenter JM, Brown KA, Diaz AN, Dockman RL, Benbow RA, Harn DA, Norberg T, Wagner JJ, Filipov NM. Delayed treatment with the immunotherapeutic LNFPIII ameliorates multiple neurological deficits in a pesticide-nerve agent prophylactic mouse model of Gulf War Illness. Neurotoxicol Teratol 2021; 87:107012. [PMID: 34256162 DOI: 10.1016/j.ntt.2021.107012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 07/03/2021] [Accepted: 07/05/2021] [Indexed: 12/20/2022]
Abstract
Residual effects of the 1990-1991 Gulf War (GW) still plague veterans 30 years later as Gulf War Illness (GWI). Thought to stem mostly from deployment-related chemical overexposures, GWI is a disease with multiple neurological symptoms with likely immunological underpinnings. Currently, GWI remains untreatable, and the long-term neurological disease manifestation is not characterized fully. The present study sought to expand and evaluate the long-term implications of prior GW chemicals exposure on neurological function 6-8 months post GWI-like symptomatology induction. Additionally, the beneficial effects of delayed treatment with the glycan immunotherapeutic lacto-N-fucopentaose III (LNFPIII) were evaluated. Male C57BL/6J mice underwent a 10-day combinational exposure (i.p.) to GW chemicals, the nerve agent prophylactic pyridostigmine bromide (PB) and the insecticide permethrin (PM; 0.7 and 200 mg/kg, respectively). Beginning 4 months after PB/PM exposure, a subset of the mice were treated twice a week until study completion with LNFPIII. Evaluation of cognition/memory, motor function, and mood was performed beginning 1 month after LNFPIII treatment initiation. Prior exposure to PB/PM produced multiple locomotor, neuromuscular, and sensorimotor deficits across several motor tests. Subtle anxiety-like behavior was also present in PB/PM mice in mood tests. Further, PB/PM-exposed mice learned at a slower rate, mostly during early phases of the learning and memory tests employed. LNFPIII treatment restored or improved many of these behaviors, particularly in motor and cognition/memory domains. Electrophysiology data collected from hippocampal slices 8 months post PB/PM exposure revealed modest aberrations in basal synaptic transmission and long-term potentiation in the dorsal or ventral hippocampus that were improved by LNFPIII treatment. Immunohistochemical analysis of tyrosine hydroxylase (TH), a dopaminergic marker, did not detect major PB/PM effects along the nigrostriatal pathway, but LNFPIII increased striatal TH. Additionally, neuroinflammatory cells were increased in PB/PM mice, an effect reduced by LNFPIII. Collectively, long-term neurobehavioral and neurobiological dysfunction associated with prior PB/PM exposure was characterized; delayed LNFPIII treatment provided multiple behavioral and biological beneficial effects in the context of GWI, highlighting its potential as a GWI therapeutic.
Collapse
Affiliation(s)
- Jessica M Carpenter
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States; Neuroscience Program, University of Georgia, Athens, GA, United States
| | - Kyle A Brown
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States; Interdisciplinary Toxicology Program, University of Georgia, Athens, GA, United States
| | - Alexa N Diaz
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States
| | - Rachel L Dockman
- Department of Microbiology, University of Georgia, Athens, GA, United States
| | - Robert A Benbow
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States
| | - Donald A Harn
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States; Center for Tropical and Emerging Infectious Diseases, University of Georgia, Athens, GA, United States
| | - Thomas Norberg
- Department of Chemistry, University of Uppsala, Uppsala, Sweden
| | - John J Wagner
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States; Neuroscience Program, University of Georgia, Athens, GA, United States; Interdisciplinary Toxicology Program, University of Georgia, Athens, GA, United States.
| | - Nikolay M Filipov
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States; Neuroscience Program, University of Georgia, Athens, GA, United States; Interdisciplinary Toxicology Program, University of Georgia, Athens, GA, United States.
| |
Collapse
|
490
|
Mancini G, Dias C, Lourenço CF, Laranjinha J, de Bem A, Ledo A. A High Fat/Cholesterol Diet Recapitulates Some Alzheimer's Disease-Like Features in Mice: Focus on Hippocampal Mitochondrial Dysfunction. J Alzheimers Dis 2021; 82:1619-1633. [PMID: 34219714 DOI: 10.3233/jad-210122] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Ample evidence from clinical and pre-clinical studies suggests mid-life hypercholesterolemia as a risk factor for developing Alzheimer's disease (AD) at a later age. Hypercholesterolemia induced by dietary habits can lead to vascular perturbations that increase the risk of developing sporadic AD. OBJECTIVE To investigate the effects of a high fat/cholesterol diet (HFCD) as a risk factor for AD by using a rodent model of AD and its correspondent control (healthy animals). METHODS We compared the effect of a HFCD in normal mice (non-transgenic mice, NTg) and the triple transgenic mouse model of AD (3xTgAD). We evaluated cognitive performance in relation to changes in oxidative metabolism and neuron-derived nitric oxide (•NO) concentration dynamics in hippocampal slices as well as histochemical staining of markers of the neurovascular unit. RESULTS In NTg, the HFCD produced only moderate hypercholesterolemia but significant decline in spatial memory was observed. A tendency for decrease in •NO production was accompanied by compromised mitochondrial function with decrease in spare respiratory capacity. In 3xTgAD mice, a robust increase in plasma cholesterol levels with the HFCD did not worsen cognitive performance but did induce compromise of mitochondrial function and significantly decreased •NO production. We found increased staining of biomarkers for astrocyte endfeet and endothelial cells in 3xTgAD hippocampi, which was further increased by the HFCD. CONCLUSION A short term (8 weeks) intervention with HFCD can produce an AD-like phenotype even in the absence of overt systemic hypercholesterolemia and highlights mitochondrial dysfunction as a link between hypercholesterolemia and sporadic AD.
Collapse
Affiliation(s)
- Gianni Mancini
- Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Candida Dias
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Catia F Lourenço
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Joao Laranjinha
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Andreza de Bem
- Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, Brazil.,Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil.,Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ana Ledo
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
491
|
Creatine Supplementation Upregulates mTORC1 Signaling and Markers of Synaptic Plasticity in the Dentate Gyrus While Ameliorating LPS-Induced Cognitive Impairment in Female Rats. Nutrients 2021; 13:nu13082758. [PMID: 34444918 PMCID: PMC8398736 DOI: 10.3390/nu13082758] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/06/2021] [Accepted: 08/10/2021] [Indexed: 11/27/2022] Open
Abstract
Mild cognitive impairment (MCI) designates the boundary area between cognitive function in natural aging and dementia, and this is viewed as a therapeutic window to prevent the occurrence of dementia. The current study investigated the neurocognitive effects of oral creatine (Cr) supplementation in young female Wistar rats that received intracerebroventricular injections of lipopolysaccharide (LPS) to mimic MCI. Neuromolecular changes within the dentate gyrus were analyzed following behavioral testing. We also investigated both neurocognitive and neuromolecular changes following Cr supplementation in the absence of LPS in young female Wistar rats to further investigate mechanisms. Interestingly, based on trial 2 of Barnes maze test, Cr supplementation ameliorated spatial learning and memory deficit induced by LPS, shown by decreased latency time and errors to reach the escape box (p < 0.0001, n = 12). Cr supplementation also attenuated recognition memory deficit induced by LPS, shown by increased amount of time taken to explore the new object (p = 0.002, n = 12) during novel object recognition testing. Within the dentate gyrus, Cr supplementation in LPS injected rats upregulated mTORC1 signaling (p = 0.026 for mTOR phosphorylation, p = 0.002 for p70S6K phosphorylation, n = 8) as well as the synapsin (p = 0.008) and PSD-95 synaptic proteins (p = 0.015), in comparisons to LPS injected rats. However, Cr supplementation failed to further enhance spatial memory and recognition memory in the absence of LPS. In conclusion, Cr ameliorates LPS-induced cognitive impairment in a rodent MCI model. Mechanistically, these phenotypic effects may, in part, be mitigated via an upregulation of mTORC1 signaling, and an enhancement in synaptogenesis in the dentate gyrus. While preliminary, these findings may inform future research investigating neurocognitive effects of Cr for MCI patients.
Collapse
|
492
|
Wang IF, Wang Y, Yang YH, Huang GJ, Tsai KJ, Shen CKJ. Activation of a hippocampal CREB-pCREB-miRNA-MEF2 axis modulates individual variation of spatial learning and memory capability. Cell Rep 2021; 36:109477. [PMID: 34348143 DOI: 10.1016/j.celrep.2021.109477] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/07/2021] [Accepted: 07/13/2021] [Indexed: 11/30/2022] Open
Abstract
Phenotypic variation is a fundamental prerequisite for cell and organism evolution by natural selection. Whereas the role of stochastic gene expression in phenotypic diversity of genetically identical cells is well studied, not much is known regarding the relationship between stochastic gene expression and individual behavioral variation in animals. We demonstrate that a specific miRNA (miR-466f-3p) is upregulated in the hippocampus of a portion of individual inbred mice upon a Morris water maze task. Significantly, miR-466f-3p positively regulates the neuron morphology, function and spatial learning, and memory capability of mice. Mechanistically, miR-466f-3p represses translation of MEF2A, a negative regulator of learning/memory. Finally, we show that varied upregulation of hippocampal miR-466f-3p results from randomized phosphorylation of hippocampal cyclic AMP (cAMP)-response element binding (CREB) in individuals. This finding of modulation of spatial learning and memory via a randomized hippocampal signaling axis upon neuronal stimulation represents a demonstration of how variation in tissue gene expression lead to varied animal behavior.
Collapse
Affiliation(s)
- I-Fang Wang
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Yihan Wang
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Yi-Hua Yang
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan; Research Center of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| | - Guo-Jen Huang
- Department and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou 33302, Taiwan
| | - Kuen-Jer Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan; Research Center of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan.
| | - Che-Kun James Shen
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan.
| |
Collapse
|
493
|
Alexander TC, Krull KR. Effects of chemotherapy for acute lymphoblastic leukemia on cognitive function in animal models of contemporary protocols: A systematic literature review. Neurosci Biobehav Rev 2021; 129:206-217. [PMID: 34352229 DOI: 10.1016/j.neubiorev.2021.07.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 10/29/2020] [Accepted: 07/28/2021] [Indexed: 11/20/2022]
Abstract
Survival rates of childhood acute lymphoblastic leukemia (ALL) have improved greatly due to advanced therapies and supportive care. Intrathecal chemotherapy replaced cranial radiation due to radiation-induced neurotoxicity and late-effects. Survivors treated with chemotherapy-only experience neurologic and cognitive problems following cessation of treatment. Very long-term cognitive outcomes remain unclear. Animal models are being generated to assess late-effects of chemotherapy on cognitive function. Although, few address juvenile models of chemotherapy-induced cognitive impairment (CICI) and developing brain, results of this review outline neurocognitive effects of chemotherapy consistent with childhood ALL therapy. Studies demonstrate deficits across cognitive domains including spatial memory, executive function, short-term memory, anxiety and depression. Inflammation, oxidative stress, excitotoxity, and other metabolic disruptions may lead to neurodegeneration associated with cognitive impairment observed in ALL survivors. Interventions directly targeting these mechanisms may prevent and/or promote recovery of cognitive function and improve long-term outcomes. Evidence suggests success of anti-inflammatory and antioxidant treatments in reducing cognitive decline. Animal models provide basis for assessing effects of chemotherapy on neurologic processes to guide future clinical investigations.
Collapse
Affiliation(s)
- Tyler C Alexander
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Kevin R Krull
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, United States; Department of Psychology, St. Jude Children's Research Hospital, Memphis, TN, United States.
| |
Collapse
|
494
|
Islam MR, Valaris S, Young MF, Haley EB, Luo R, Bond SF, Mazuera S, Kitchen RR, Caldarone BJ, Bettio LEB, Christie BR, Schmider AB, Soberman RJ, Besnard A, Jedrychowski MP, Kim H, Tu H, Kim E, Choi SH, Tanzi RE, Spiegelman BM, Wrann CD. Exercise hormone irisin is a critical regulator of cognitive function. Nat Metab 2021; 3:1058-1070. [PMID: 34417591 PMCID: PMC10317538 DOI: 10.1038/s42255-021-00438-z] [Citation(s) in RCA: 192] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 07/13/2021] [Indexed: 02/07/2023]
Abstract
Identifying secreted mediators that drive the cognitive benefits of exercise holds great promise for the treatment of cognitive decline in ageing or Alzheimer's disease (AD). Here, we show that irisin, the cleaved and circulating form of the exercise-induced membrane protein FNDC5, is sufficient to confer the benefits of exercise on cognitive function. Genetic deletion of Fndc5/irisin (global Fndc5 knock-out (KO) mice; F5KO) impairs cognitive function in exercise, ageing and AD. Diminished pattern separation in F5KO mice can be rescued by delivering irisin directly into the dentate gyrus, suggesting that irisin is the active moiety. In F5KO mice, adult-born neurons in the dentate gyrus are morphologically, transcriptionally and functionally abnormal. Importantly, elevation of circulating irisin levels by peripheral delivery of irisin via adeno-associated viral overexpression in the liver results in enrichment of central irisin and is sufficient to improve both the cognitive deficit and neuropathology in AD mouse models. Irisin is a crucial regulator of the cognitive benefits of exercise and is a potential therapeutic agent for treating cognitive disorders including AD.
Collapse
Affiliation(s)
- Mohammad R Islam
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sophia Valaris
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael F Young
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Erin B Haley
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Renhao Luo
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sabrina F Bond
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Program in Behavioral Neuroscience, Northeastern University, Boston, MA, USA
| | - Sofia Mazuera
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Program in Behavioral Neuroscience, Northeastern University, Boston, MA, USA
| | - Robert R Kitchen
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Barbara J Caldarone
- Harvard NeuroDiscovery Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Luis E B Bettio
- Division of Medical Sciences, University of Victoria, Victoria, British Colombia, Canada
| | - Brian R Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Colombia, Canada
| | - Angela B Schmider
- Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Roy J Soberman
- Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Antoine Besnard
- Center for Regenerative Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Mark P Jedrychowski
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Hyeonwoo Kim
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Hua Tu
- LakePharma, San Carlos, CA, USA
| | - Eunhee Kim
- MassGeneral Institute for Neurodegenerative Disease, Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Se Hoon Choi
- MassGeneral Institute for Neurodegenerative Disease, Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Rudolph E Tanzi
- MassGeneral Institute for Neurodegenerative Disease, Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Christiane D Wrann
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
495
|
Głuch-Lutwin M, Sałaciak K, Gawalska A, Jamrozik M, Sniecikowska J, Newman-Tancredi A, Kołaczkowski M, Pytka K. The selective 5-HT 1A receptor biased agonists, F15599 and F13714, show antidepressant-like properties after a single administration in the mouse model of unpredictable chronic mild stress. Psychopharmacology (Berl) 2021; 238:2249-2260. [PMID: 33973045 PMCID: PMC8292235 DOI: 10.1007/s00213-021-05849-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 04/12/2021] [Indexed: 01/12/2023]
Abstract
RATIONALE The prevalence of depression is ever-increasing throughout the population. However, available treatments are ineffective in around one-third of patients and there is a need for more effective and safer drugs. OBJECTIVES The antidepressant-like and procognitive effects of the "biased agonists" F15599 (also known as NLX-101) which preferentially targets postsynaptic 5-HT1A receptors and F13714, which targets 5-HT1A autoreceptors, were investigated in mice. METHODS Antidepressant-like properties of the compounds and their effect on cognitive functions were assessed using the forced swim test (FST) and the novel object recognition (NOR), respectively. Next, we induced a depressive-like state by an unpredictable chronic mild stress (UCMS) procedure to test the compounds' activity in the depression model, followed by measures of sucrose preference, FST, and locomotor activity. Levels of phosphorylated cyclic AMP response element-binding protein (p-CREB) and phosphorylated extracellular signal-regulated kinase (p-ERK1/2) were also determined. RESULTS F15599 reduced immobility time in the FST over a wider dose-range (2 to 16 mg/kg po) than F13714 (2 and 4 mg/kg po), suggesting accentuated antidepressant-like properties in mice. F15599 did not disrupt long-term memory consolidation in the NOR at any dose tested, while F13714 impaired memory formation, notably at higher doses (4-16 mg/kg). In UCMS mice, a single administration of F15599 and F13714 was sufficient to robustly normalize depressive-like behavior in the FST but did not rescue disrupted sucrose preference. Both F15599 and F13714 rescued cortical and hippocampal deficits in p-ERK1/2 levels of UCMS mice but did not influence the p-CREB levels. CONCLUSIONS Our studies showed that 5-HT1A receptor biased agonists such as F13714 and especially F15599, due to its less pronounced side effects, might have potential as fast-acting antidepressants.
Collapse
Affiliation(s)
- Monika Głuch-Lutwin
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Kinga Sałaciak
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Alicja Gawalska
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Marek Jamrozik
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Joanna Sniecikowska
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | | | - Marcin Kołaczkowski
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Karolina Pytka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
496
|
Arora S, Dharavath RN, Bansal Y, Bishnoi M, Kondepudi KK, Chopra K. Neurobehavioral alterations in a mouse model of chronic partial sleep deprivation. Metab Brain Dis 2021; 36:1315-1330. [PMID: 33740181 DOI: 10.1007/s11011-021-00693-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 02/12/2021] [Indexed: 12/23/2022]
Abstract
The night shift paradigm induces a state of chronic partial sleep deprivation (CPSD) and enhances the vulnerability to neuronal dysfunction. However, the specific neuronal impact of CPSD has not been thoroughly explored to date. In the current study, the night shift condition was mimicked in female Swiss albino mice. The classical sleep deprivation model, i.e., Modified Multiple Platform (MMP) method, was used for 8 h/day from Monday to Friday with Saturday and Sunday as a weekend off for nine weeks. Following nine weeks of night shift schedule, their neurobehavioral profile and physiological parameters were assessed along with the activity of the mitochondrial complexes, oxidative stress, serotonin levels, and inflammatory markers in the brain. Mice showed an overall hyperactive behavioral profile including hyperlocomotion, aggression, and stereotyped behavior accompanied by decreased activity of mitochondrial enzymes and serotonin levels, increased oxidative stress and inflammatory markers in whole brain homogenates. Collectively, the study points towards the occurrence of a hyperactive behavioral profile akin to mania and psychosis as a potential consequence of CPSD.
Collapse
Affiliation(s)
- Shiyana Arora
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Sector 14, 160014, Chandigarh, India
| | - Ravinder Naik Dharavath
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Sector 14, 160014, Chandigarh, India
| | - Yashika Bansal
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Sector 14, 160014, Chandigarh, India
| | - Mahendra Bishnoi
- Food and Nutritional Biotechnology Laboratory, National Agri-Food Biotechnology Institute, SAS Nagar, Punjab, 140306, India
| | - Kanthi Kiran Kondepudi
- Food and Nutritional Biotechnology Laboratory, National Agri-Food Biotechnology Institute, SAS Nagar, Punjab, 140306, India
| | - Kanwaljit Chopra
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Sector 14, 160014, Chandigarh, India.
| |
Collapse
|
497
|
Assessment of Antidepressant-like, Anxiolytic Effects and Impact on Memory of Pimpinella anisum L. Total Extract on Swiss Albino Mice. PLANTS 2021; 10:plants10081573. [PMID: 34451620 PMCID: PMC8399924 DOI: 10.3390/plants10081573] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 12/20/2022]
Abstract
Mental disorders are psychological symptoms that impact multiple areas of an individual’s life. Depression and anxiety are chronic illnesses described as the most prevalent stress-related mood disorders that cause injury and early death. In Morocco, Anise “Pimpinella anisum L.” is one of the most traditionally used condiment plants, which has long been used to cure various illnesses and in phytotherapy. The present study was designed to investigate the antidepressant, anxiolytic, and memory impact of the total extract of Pimpinella anisum (PATE) at the doses of 100 and 200 mg/kg, using the Forced Swimming Test (FST), Tail Suspension Test (TST), Open Field Test (OFT), and Light–Dark Box Test (LDBT) as an experimental paradigm of anxiety and depression, and Novel Object Recognition Test (NORT) and the Morris Water Maze Test (MWMT) as memory tests on Swiss albino mice. The tests were carried out on the 1st, 7th, 14th, and the 21st days of the study, and the extract groups were compared with normal controls and positive controls (receiving bromazepam and paroxetine at the doses of 1 mg/kg and 11.5 mg/kg for anxiety and depression, respectively). The daily oral gavage of the mice by the PATE induced a significant anxiolytic and antidepressant-like effect by shortening immobility time and decreasing downtime in the different tests. PATE at both doses was shown to have no impact on memory following the NORT and MWM tests. Different compounds, such as gallic acid, catechin, chlorogenic acid, caffeic acid, oleuropein, p-coumaric acid, trans-4-hydroxy-3-methoxycinnamic acid, myricetin, and quercetin, were identified during the phytochemical analysis carried out using HPLC analysis. This research supports and promotes the extract’s traditional use, suggesting its use as a phytomedicine against depression and anxiety, and calls for further research to clarify its mode of action.
Collapse
|
498
|
Ehinger Y, Zhang Z, Phamluong K, Soneja D, Shokat KM, Ron D. Brain-specific inhibition of mTORC1 eliminates side effects resulting from mTORC1 blockade in the periphery and reduces alcohol intake in mice. Nat Commun 2021; 12:4407. [PMID: 34315870 PMCID: PMC8316332 DOI: 10.1038/s41467-021-24567-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 06/23/2021] [Indexed: 12/11/2022] Open
Abstract
Alcohol Use Disorder (AUD) affects a large portion of the population. Unfortunately, efficacious medications to treat the disease are limited. Studies in rodents suggest that mTORC1 plays a crucial role in mechanisms underlying phenotypes such as heavy alcohol intake, habit, and relapse. Thus, mTORC1 inhibitors, which are used in the clinic, are promising therapeutic agents to treat AUD. However, chronic inhibition of mTORC1 in the periphery produces undesirable side effects, which limit their potential use for the treatment of AUD. To overcome these limitations, we designed a binary drug strategy in which male mice were treated with the mTORC1 inhibitor RapaLink-1 together with a small molecule (RapaBlock) to protect mTORC1 activity in the periphery. We show that whereas RapaLink-1 administration blocked mTORC1 activation in the liver, RapaBlock abolished the inhibitory action of Rapalink-1. RapaBlock also prevented the adverse side effects produced by chronic inhibition of mTORC1. Importantly, co-administration of RapaLink-1 and RapaBlock inhibited alcohol-dependent mTORC1 activation in the nucleus accumbens and attenuated alcohol seeking and drinking.
Collapse
Affiliation(s)
- Yann Ehinger
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Ziyang Zhang
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, CA, USA
| | - Khanhky Phamluong
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Drishti Soneja
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Kevan M Shokat
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, CA, USA
| | - Dorit Ron
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
499
|
Zhang R, Wang J, Huang L, Wang TJ, Huang Y, Li Z, He J, Sun C, Wang J, Chen X, Wang J. The pros and cons of motor, memory, and emotion-related behavioral tests in the mouse traumatic brain injury model. Neurol Res 2021; 44:65-89. [PMID: 34308784 DOI: 10.1080/01616412.2021.1956290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Traumatic brain injury (TBI) is a medical emergency with high morbidity and mortality. Motor, memory, and emotion-related deficits are common symptoms following TBI, yet treatment is very limited. To develop new drugs and find new therapeutic avenues, a wide variety of TBI models have been established to mimic the heterogeneity of TBI. In this regard, along with histologic measures, behavioral functional outcomes provide valuable insight into the underlying neuropathology and guide neurorehabilitation efforts for neuropsychiatric impairment after TBI. Development, characterization, and application of behavioral tests that can assess functional neurologic deficits are essential to the development of translational therapies. This comprehensive review aims to summarize 19 common behavioral tests from three aspects (motor, memory, and emotion-related) that are associated with TBI pathology. Discussion covers the apparatus, the test steps, the evaluation indexes, data collection and analysis, animal performance and applications, advantages and disadvantages as well as precautions to eliminate bias wherever possible. We discussed recent studies on TBI-related preconditioning, biomarkers, and optimized behavioral protocols. The neuropsychologic tests employed in clinics were correlated with those used in mouse TBI models. In summary, this review provides a comprehensive, up-to-date reference for TBI researchers to choose the right neurobehavioral protocol according to the research objectives of their translational investigation.
Collapse
Affiliation(s)
- Ruoyu Zhang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Junming Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Leo Huang
- Department of Psychology, University of Toronto, Toronto, Canada
| | - Tom J Wang
- Winston Churchill High School, Potomac, Maryland, USA
| | - Yinrou Huang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zefu Li
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jinxin He
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Chen Sun
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jing Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xuemei Chen
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jian Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
500
|
de Sousa LP, Ribeiro-Gomes FL, de Almeida RF, Souza TME, Werneck GL, Souza DO, Daniel-Ribeiro CT. Immune system challenge improves recognition memory and reverses malaria-induced cognitive impairment in mice. Sci Rep 2021; 11:14857. [PMID: 34290279 PMCID: PMC8295320 DOI: 10.1038/s41598-021-94167-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/05/2021] [Indexed: 11/09/2022] Open
Abstract
The immune system plays a role in the maintenance of healthy neurocognitive function. Different patterns of immune response triggered by distinct stimuli may affect nervous functions through regulatory or deregulatory signals, depending on the properties of the exogenous immunogens. Here, we investigate the effect of immune stimulation on cognitive-behavioural parameters in healthy mice and its impact on cognitive sequelae resulting from non-severe experimental malaria. We show that immune modulation induced by a specific combination of immune stimuli that induce a type 2 immune response can enhance long-term recognition memory in healthy adult mice subjected to novel object recognition task (NORT) and reverse a lack of recognition ability in NORT and anxiety-like behaviour in a light/dark task that result from a single episode of mild Plasmodium berghei ANKA malaria. Our findings suggest a potential use of immunogens for boosting and recovering recognition memory that may be impaired by chronic and infectious diseases and by the effects of ageing.
Collapse
Affiliation(s)
- Luciana Pereira de Sousa
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) of Fundação Oswaldo Cruz (Fiocruz) and of Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Fiocruz. Av. Brasil 4365, Manguinhos, Rio de Janeiro, RJ, CEP 2104-360, Brazil
| | - Flávia Lima Ribeiro-Gomes
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) of Fundação Oswaldo Cruz (Fiocruz) and of Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Fiocruz. Av. Brasil 4365, Manguinhos, Rio de Janeiro, RJ, CEP 2104-360, Brazil
| | - Roberto Farina de Almeida
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Minas Gerais, Brazil
| | - Tadeu Mello E Souza
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Guilherme Loureiro Werneck
- Departamento de Epidemiologia of Instituto de Medicina Social, Universidade do Estado do Rio de Janeiro and Instituto de Estudos de Saúde Coletiva da Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diogo Onofre Souza
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Cláudio Tadeu Daniel-Ribeiro
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) of Fundação Oswaldo Cruz (Fiocruz) and of Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Fiocruz. Av. Brasil 4365, Manguinhos, Rio de Janeiro, RJ, CEP 2104-360, Brazil.
| |
Collapse
|