1
|
Serebrovska Z, Xi L, Fedoriuk M, Dosenko V, Shysh A, Khetsuriani M, Porkhalo D, Savchenko A, Goncharov S, Utko N, Virko S, Kholin V, Egorov E, Koval R, Maksymchuk O. Intermittent hypoxia-hyperoxia training ameliorates cognitive impairment and neuroinflammation in a rat model of Alzheimer's disease. Brain Res 2025; 1847:149301. [PMID: 39476996 DOI: 10.1016/j.brainres.2024.149301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 10/02/2024] [Accepted: 10/26/2024] [Indexed: 11/15/2024]
Abstract
Alzheimer's disease (AD), characterized by severe and progressive cognitive decline, stands as one of the most prevalent and devastating forms of dementia. Based on our recent findings showing intermittent hypoxic conditioning improved neuronal function in patients with mild cognitive impairment, the present study aimed at investigating whether the neuroprotective effects of intermittent hypoxia can be replicated in a rat model of AD, which allows us to explore the underlying cellular mechanisms involving neuroinflammation, hypoxia inducible factor 1α (HIF1α), and cytochrome P450 family 2 subfamily E member 1 (CYP2E1). Forty-one adult male Wistar rats were randomly assigned to three groups: 1) Control group: received intracerebroventricular (ICV) injection of saline; 2) STZ group: received ICV injection of streptozotocin (STZ) to induce AD-like pathology; and 3) STZ + IHHT group received ICV injection of STZ as well as 15 daily sessions of intermittent hypoxia-hyperoxia training (IHHT). We observed that ICV injection of STZ inhibited spatial learning and memory in the rats assessed with Morris Water Maze test. The cognitive function declines were accompanied by increased expression of amyloid β peptide (Aβ), HIF1α, CYP2E1, and TNFα in hippocampus. Interestingly, IHHT significantly restored the STZ-induced cognitive dysfunction, while reduced expression of Aβ, CYP2E1, HIF1α and TNFα. We conclude that IHHT with mild hypoxia-hyperoxia can enhance spatial learning and memory and reduce the AD-like pathologic changes in rats. The neuroprotective outcome of IHHT may be related to anti-inflammatory effects in hippocampus.
Collapse
Affiliation(s)
- Zoya Serebrovska
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv 01601, Ukraine.
| | - Lei Xi
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298-0204, USA.
| | - Mykhailo Fedoriuk
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv 01601, Ukraine
| | - Victor Dosenko
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv 01601, Ukraine
| | - Angela Shysh
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv 01601, Ukraine
| | - Michael Khetsuriani
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv 01601, Ukraine
| | - Denys Porkhalo
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv 01601, Ukraine
| | - Anton Savchenko
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv 01601, Ukraine
| | - Serhii Goncharov
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv 01601, Ukraine
| | - Natalie Utko
- Chebotarev Institute of Gerontology, National Academy of Sciences of Ukraine, Kyiv 04114, Ukraine
| | - Sergii Virko
- Lashkariov Institute of Semiconductor Physics, National Academy of Sciences, Kyiv 02000, Ukraine
| | - Victor Kholin
- Chebotarev Institute of Gerontology, National Academy of Sciences of Ukraine, Kyiv 04114, Ukraine
| | - Egor Egorov
- Institute for Preventive and Antiaging Medicine, Berlin 10789, Germany
| | - Roman Koval
- National Cancer Institute, Kyiv 03022, Ukraine
| | - Oksana Maksymchuk
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv 03143, Ukraine
| |
Collapse
|
2
|
Wei RM, Zhang MY, Fang SK, Liu GX, Hu F, Li XY, Zhang KX, Zhang JY, Liu XC, Zhang YM, Chen GH. Melatonin attenuates intermittent hypoxia-induced cognitive impairment in aged mice: The role of inflammation and synaptic plasticity. Psychoneuroendocrinology 2025; 171:107210. [PMID: 39378690 DOI: 10.1016/j.psyneuen.2024.107210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 09/08/2024] [Accepted: 10/02/2024] [Indexed: 10/10/2024]
Abstract
Intermittent hypoxia (IH), a major pathophysiologic alteration in obstructive sleep apnea syndrome (OSAS), is an important contributor to cognitive impairment. Increasing research suggests that melatonin has anti-inflammatory properties and improves functions related to synaptic plasticity. However, it is unclear whether melatonin has a protective effect against OSAS-induced cognitive dysfunction in aged individuals and the involved mechanisms are also unclear. Therefore, in the study, the effects of exposure to IH alone and IH in combination with daily melatonin treatment were investigated in C57BL/6 J mice aged 18 months. Assessment of the cognitive ability of mice in a Morris water maze showed that melatonin attenuated IH-induced impairment of learning and memory in aged mice. Enzyme-linked immunosorbent assay, polymerase chain reaction, and western blotting molecular techniques showed that melatonin treatment reduced the levels of the proinflammatory cytokines, interleukin-1β, interleukin-6, and tumor necrosis factor-α, decreased the levels of NOD-like receptor thermal protein domain associated protein 3 and nuclear factor kappa-B, lowered the levels of ionized calcium-binding adapter molecule 1 and glial fibrillary acidic protein, and increased the levels of the synaptic proteins, activity-regulated cytoskeleton-associated protein, growth-associated protein-43, postsynaptic density protein 95, and synaptophysin in IH-exposed mice. Moreover, electrophysiological results showed that melatonin ameliorated the decline in long-term potentiation induced by IH. The results suggest that melatonin can ameliorate IH-induced cognitive deficits by inhibiting neuroinflammation and improving synaptic plasticity in aged mice.
Collapse
Affiliation(s)
- Ru-Meng Wei
- Department of Neurology (sleep disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| | - Meng-Ying Zhang
- Department of Anesthesiology, the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| | - Shi-Kun Fang
- Department of Neurology (sleep disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| | - Gao-Xia Liu
- Department of Neurology (sleep disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| | - Fei Hu
- Department of Neurology (sleep disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| | - Xue-Yan Li
- Department of Neurology (sleep disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| | - Kai-Xuan Zhang
- Department of Neurology (sleep disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| | - Jing-Ya Zhang
- Department of Neurology (sleep disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| | - Xue-Chun Liu
- Department of Neurology, the Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China.
| | - Yue-Ming Zhang
- Department of Neurology (sleep disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| | - Gui-Hai Chen
- Department of Neurology (sleep disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| |
Collapse
|
3
|
Regalbuto S, Zangaglia R, Valentino F, Todisco M, Pacchetti C, Cotta Ramusino M, Mazzacane F, Picascia M, Arceri S, Malomo G, Capriglia E, Spelta L, Rubino A, Pisani A, Terzaghi M. Clinical correlates of obstructive sleep apnoea in idiopathic normal pressure hydrocephalus. Eur J Neurol 2024; 31:e16448. [PMID: 39207116 PMCID: PMC11555027 DOI: 10.1111/ene.16448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/30/2024] [Accepted: 08/04/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND AND PURPOSE The pathogenesis of idiopathic normal pressure hydrocephalus (iNPH) remains controversial. Limited studies have indicated a high prevalence of obstructive sleep apnoea (OSA) amongst iNPH patients. The aim was to investigate the clinical correlates of OSA in iNPH patients. METHODS In this cross-sectional observational study, consecutive iNPH patients were prospectively enrolled. Evaluations included the iNPH Rating Scale, the Movement Disorder Society Unified Parkinson's Disease Rating Scale part III, the time and number of steps to walk 10 m, the Epworth Sleepiness Scale, the Pittsburgh Sleep Quality Index, a complete neuropsychological evaluation, 3-T brain MRI, full-night video-polysomnography, tap test and cerebrospinal fluid (CSF) neurodegeneration biomarkers. RESULTS Fifty-one patients were screened, of whom 38 met the inclusion criteria. Amongst the recruited patients, 19/38 (50%) exhibited OSA, with 12/19 (63.2%) presenting moderate to severe disorder. OSA+ iNPH patients required more time (p = 0.02) and more steps (p = 0.04) to complete the 10-m walking test, had lower scores on the gait subitem of the iNPH Rating Scale (p = 0.04) and demonstrated poorer performance on specific neuropsychological tests (Rey Auditory Verbal Learning Test immediate recall, p = 0.03, and Rey-Osterrieth Complex Figure, p = 0.01). Additionally, OSA+ iNPH patients had higher levels of total tau (p = 0.02) and phospho-tau (p = 0.03) in their CSF but no statistically significant differences in beta-amyloid (1-42) levels compared to OSA- iNPH patients. CONCLUSION Obstructive sleep apnoea is highly prevalent in iNPH patients, particularly at moderate to severe levels. OSA is associated with worse motor and cognitive performance in iNPH. The CSF neurodegeneration biomarker profile observed in OSA+ iNPH patients may reflect OSA-induced impairment of cerebral fluid dynamics.
Collapse
Affiliation(s)
- Simone Regalbuto
- IRCCS Mondino FoundationNational Neurological InstitutePaviaItaly
| | | | | | | | | | - Matteo Cotta Ramusino
- IRCCS Mondino FoundationNational Neurological InstitutePaviaItaly
- Department of Brain and Behavioural SciencesUniversity of PaviaPaviaItaly
| | - Federico Mazzacane
- Department of Brain and Behavioural SciencesUniversity of PaviaPaviaItaly
| | - Marta Picascia
- IRCCS Mondino FoundationNational Neurological InstitutePaviaItaly
| | | | - Gaetano Malomo
- Department of Brain and Behavioural SciencesUniversity of PaviaPaviaItaly
| | - Elena Capriglia
- Department of Brain and Behavioural SciencesUniversity of PaviaPaviaItaly
| | - Laura Spelta
- IRCCS Mondino FoundationNational Neurological InstitutePaviaItaly
| | - Annalisa Rubino
- IRCCS Mondino FoundationNational Neurological InstitutePaviaItaly
| | - Antonio Pisani
- IRCCS Mondino FoundationNational Neurological InstitutePaviaItaly
- Department of Brain and Behavioural SciencesUniversity of PaviaPaviaItaly
| | - Michele Terzaghi
- IRCCS Mondino FoundationNational Neurological InstitutePaviaItaly
- Department of Brain and Behavioural SciencesUniversity of PaviaPaviaItaly
| |
Collapse
|
4
|
Zhang C, Wang Y, Li M, Niu P, Li S, Hu Z, Shi C, Li Y. Phase-Amplitude Coupling in Theta and Beta Bands: A Potential Electrophysiological Marker for Obstructive Sleep Apnea. Nat Sci Sleep 2024; 16:1469-1482. [PMID: 39323903 PMCID: PMC11423842 DOI: 10.2147/nss.s470617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024] Open
Abstract
Background Phase-amplitude coupling (PAC) between the phase of low-frequency signals and the amplitude of high-frequency activities plays many physiological roles and is involved in the pathological processed of various neurological disorders. However, how low-frequency and high-frequency neural oscillations or information synchronization activities change under chronic central hypoxia in OSA patients and whether these changes are closely associated with OSA remains largely unexplored. This study arm to elucidate the long-term consequences of OSA-related oxygen deprivation on central nervous system function. Methods : We screened 521 patients who were clinically suspected of having OSA at our neurology and sleep centers. Through polysomnography (PSG) and other clinical examinations, 103 patients were ultimately included in the study and classified into mild, moderate, and severe OSA groups based on the severity of hypoxia determined by PSG. We utilized the phase-amplitude coupling (PAC) method to analyze the modulation index (MI) trends between different frequency bands during NREM (N1/N2/N3), REM, and wakefulness stages in OSA patients with varying severity levels. We also examined the correlation between the MI index and OSA hypoxia indices. Results Apart from reduced N2 sleep duration and increased microarousal index, the sleep architecture remained largely unchanged among OSA patients with varying severity levels. Compared to the mild OSA group, patients with moderate and severe OSA exhibited higher MI values of PAC in the low-frequency theta phase and high-frequency beta amplitude in the frontal and occipital regions during N1 sleep and wakefulness. No significant differences in the MI of phase-amplitude coupling were observed during N2/3 and REM sleep. Moreover, the MI of phase-amplitude coupling in theta and beta bands positively correlated with hypoxia-related indices, including the apnea-hypopnea index (AHI) and oxygenation desaturation index (ODI), and the percentage of oxygen saturation below 90% (SaO2<90%). Conclusion OSA patients demonstrated increased MI values of theta phase and beta amplitude in the frontal and occipital regions during N1 sleep and wakefulness. This suggests that cortical coupling is prevalent and exhibits sleep-stage-specific patterns in OSA. Theta-beta PAC during N1 and wakefulness was positively correlated with hypoxia-related indices, suggesting a potential relationship between these neural oscillations and OSA severity. The present study provides new insights into the relationship between neural oscillations and respiratory hypoxia in OSA patients.
Collapse
Affiliation(s)
- Chan Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450000, People’s Republic of China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, Henan, People’s Republic of China
- Henan Neurological Function Detection and Regulation Center, Zhengzhou, Henan, 450000, People’s Republic of China
| | - Yanhui Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450000, People’s Republic of China
- Henan Neurological Function Detection and Regulation Center, Zhengzhou, Henan, 450000, People’s Republic of China
- The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Mengjie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450000, People’s Republic of China
- The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Pengpeng Niu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450000, People’s Republic of China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, Henan, People’s Republic of China
| | - Shuo Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450000, People’s Republic of China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, Henan, People’s Republic of China
| | - Zhuopeng Hu
- The First Bethune Clinical Medical College of Ji Lin University, Changchun, Jilin, People’s Republic of China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450000, People’s Republic of China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, Henan, People’s Republic of China
| | - Yusheng Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450000, People’s Republic of China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, Henan, People’s Republic of China
- Henan Neurological Function Detection and Regulation Center, Zhengzhou, Henan, 450000, People’s Republic of China
| |
Collapse
|
5
|
Doody NE, Smith NJ, Akam EC, Askew GN, Kwok JCF, Ichiyama RM. Differential expression of genes in the RhoA/ROCK pathway in the hippocampus and cortex following intermittent hypoxia and high-intensity interval training. J Neurophysiol 2024; 132:531-543. [PMID: 38985935 PMCID: PMC11427053 DOI: 10.1152/jn.00422.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 06/13/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024] Open
Abstract
Structural neuroplasticity such as neurite extension and dendritic spine dynamics is enhanced by brain-derived neurotrophic factor (BDNF) and impaired by types of inhibitory molecules that induce growth cone collapse and actin depolymerization, for example, myelin-associated inhibitors, chondroitin sulfate proteoglycans, and negative guidance molecules. These inhibitory molecules can activate RhoA/rho-associated coiled-coil containing protein kinase (ROCK) signaling (known to restrict structural plasticity). Intermittent hypoxia (IH) and high-intensity interval training (HIIT) are known to upregulate BDNF that is associated with improvements in learning and memory and greater functional recovery following neural insults. We investigated whether the RhoA/ROCK signaling pathway is also modulated by IH and HIIT in the hippocampus, cortex, and lumbar spinal cord of male Wistar rats. The gene expression of 25 RhoA/ROCK signaling pathway components was determined following IH, HIIT, or IH combined with HIIT (30 min/day, 5 days/wk, 6 wk). IH included 10 3-min bouts that alternated between hypoxia (15% O2) and normoxia. HIIT included 10 3-min bouts alternating between treadmill speeds of 50 cm·s-1 and 15 cm·s-1. In the hippocampus, IH and HIIT significantly downregulated Acan and NgR2 mRNA that are involved in the inhibition of neuroplasticity. However, IH and IH + HIIT significantly upregulated Lingo-1 and NgR3 in the cortex. This is the first time IH and HIIT have been linked to the modulation of plasticity-inhibiting pathways. These results provide a fundamental step toward elucidating the interplay between the neurotrophic and inhibitory mechanisms involved in experience-driven neural plasticity that will aid in optimizing physiological interventions for the treatment of cognitive decline or neurorehabilitation.NEW & NOTEWORTHY Intermittent hypoxia (IH) and high-intensity interval training (HIIT) enhance neuroplasticity and upregulate neurotrophic factors in the central nervous system (CNS). We provide evidence that IH and IH + HIIT also have the capacity to regulate genes involved in the RhoA/ROCK signaling pathway that is known to restrict structural plasticity in the CNS. This provides a new mechanistic insight into how these interventions may enhance hippocampal-related plasticity and facilitate learning, memory, and neuroregeneration.
Collapse
Affiliation(s)
- Natalie E Doody
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, United Kingdom
| | - Nicole J Smith
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Elizabeth C Akam
- School of Sport, Exercise, and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Graham N Askew
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Jessica C F Kwok
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
- Institute of Experimental Medicine, The Czech Academy of Sciences, Prague 4, Czech Republic
| | - Ronaldo M Ichiyama
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
6
|
He Y, Dong N, Wang X, Lv RJ, Yu Q, Yue HM. Obstructive sleep apnea affects cognition: dual effects of intermittent hypoxia on neurons. Sleep Breath 2024; 28:1051-1065. [PMID: 38308748 DOI: 10.1007/s11325-024-03001-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 02/05/2024]
Abstract
Obstructive sleep apnea (OSA) is a common respiratory disorder. Multiple organs, especially the central nervous system (CNS), are damaged, and dysfunctional when intermittent hypoxia (IH) occurs during sleep for a long time. The quality of life of individuals with OSA is significantly impacted by cognitive decline, which also escalates the financial strain on their families. Consequently, the development of novel therapies becomes imperative. IH induces oxidative stress, endoplasmic reticulum stress, iron deposition, and neuroinflammation in neurons. Synaptic dysfunction, reactive gliosis, apoptosis, neuroinflammation, and inhibition of neurogenesis can lead to learning and long-term memory impairment. In addition to nerve injury, the role of IH in neuroprotection was also explored. While causing neuron damage, IH activates the neuronal self-repairing mechanism by regulating antioxidant capacity and preventing toxic protein deposition. By stimulating the proliferation and differentiation of neural stem cells (NSCs), IH has the potential to enhance the ratio of neonatal neurons and counteract the decline in neuron numbers. This review emphasizes the perspectives and opportunities for the neuroprotective effects of IH and informs novel insights and therapeutic strategies in OSA.
Collapse
Affiliation(s)
- Yao He
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Na Dong
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xiao Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Ren-Jun Lv
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Qin Yu
- Department of Respiratory and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| | - Hong-Mei Yue
- Department of Respiratory and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
7
|
Yu Z, Teng Y, Yang J, Yang L. The role of exosomes in adult neurogenesis: implications for neurodegenerative diseases. Neural Regen Res 2024; 19:282-288. [PMID: 37488879 PMCID: PMC10503605 DOI: 10.4103/1673-5374.379036] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/12/2023] [Accepted: 05/16/2023] [Indexed: 07/26/2023] Open
Abstract
Exosomes are cup-shaped extracellular vesicles with a lipid bilayer that is approximately 30 to 200 nm in thickness. Exosomes are widely distributed in a range of body fluids, including urine, blood, milk, and saliva. Exosomes exert biological function by transporting factors between different cells and by regulating biological pathways in recipient cells. As an important form of intercellular communication, exosomes are increasingly being investigated due to their ability to transfer bioactive molecules such as lipids, proteins, mRNAs, and microRNAs between cells, and because they can regulate physiological and pathological processes in the central nervous system. Adult neurogenesis is a multistage process by which new neurons are generated and migrate to be integrated into existing neuronal circuits. In the adult brain, neurogenesis is mainly localized in two specialized niches: the subventricular zone adjacent to the lateral ventricles and the subgranular zone of the dentate gyrus. An increasing body of evidence indicates that adult neurogenesis is tightly controlled by environmental conditions with the niches. In recent studies, exosomes released from different sources of cells were shown to play an active role in regulating neurogenesis both in vitro and in vivo, thereby participating in the progression of neurodegenerative disorders in patients and in various disease models. Here, we provide a state-of-the-art synopsis of existing research that aimed to identify the diverse components of exosome cargoes and elucidate the therapeutic potential of exosomal contents in the regulation of neurogenesis in several neurodegenerative diseases. We emphasize that exosomal cargoes could serve as a potential biomarker to monitor functional neurogenesis in adults. In addition, exosomes can also be considered as a novel therapeutic approach to treat various neurodegenerative disorders by improving endogenous neurogenesis to mitigate neuronal loss in the central nervous system.
Collapse
Affiliation(s)
- Zhuoyang Yu
- Institute of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Yan Teng
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Jing Yang
- Institute of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Lu Yang
- Institute of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| |
Collapse
|
8
|
Liu Y, Tan J, Miao Y, Zhang Q. Neurogenesis, A Potential Target for Intermittent Hypoxia Leading to Cognitive Decline. Curr Stem Cell Res Ther 2024; 19:63-70. [PMID: 37005547 DOI: 10.2174/1574888x18666230330083206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 04/04/2023]
Abstract
As a sleep breathing disorder, characterized by intermittent hypoxia (IH) and Obstructive sleep apnea (OSA), is believed to decrease the cognitive function of patients. Many factors are thought to be responsible for cognitive decline in OSA patients. Neurogenesis, a process by which neural stem cells (NSCs) differentiate into new neurons in the brain, is a major determinant affecting cognitive function. However, there is no clear relationship between IH or OSA and neurogenesis. In recent years, increasing numbers of studies on IH and neurogenesis are documented. Therefore, this review summarizes the effects of IH on neurogenesis; then discusses the influencing factors that may cause these effects and the potential signaling pathways that may exist. Finally, based on this impact, we discuss potential methods and future directions for improving cognition.
Collapse
Affiliation(s)
- Yuxing Liu
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Jin Tan
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Yuyang Miao
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Qiang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| |
Collapse
|
9
|
André C, Kuhn E, Rehel S, Ourry V, Demeilliez-Servouin S, Palix C, Felisatti F, Champetier P, Dautricourt S, Yushkevich P, Vivien D, de La Sayette V, Chételat G, de Flores R, Rauchs G. Association of Sleep-Disordered Breathing and Medial Temporal Lobe Atrophy in Cognitively Unimpaired Amyloid-Positive Older Adults. Neurology 2023; 101:e370-e385. [PMID: 37258299 PMCID: PMC10435067 DOI: 10.1212/wnl.0000000000207421] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 04/03/2023] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Sleep disordered breathing (SDB) has been related to amyloid deposition and an increased dementia risk. However, how SDB relates to medial temporal lobe neurodegeneration and subsequent episodic memory impairment is unclear. Our objective was to investigate the impact of amyloid positivity on the associations between SDB severity, medial temporal lobe subregions, and episodic memory performance in cognitively unimpaired older adults. METHODS Data were acquired between 2016 and 2020 in the context of the Age-Well randomized controlled trial of the Medit-Aging European project. Participants older than 65 years who were free of neurologic, psychiatric, or chronic medical diseases were recruited from the community. They completed a neuropsychological evaluation, in-home polysomnography, a Florbetapir PET, and an MRI, including a specific high-resolution assessment of the medial temporal lobe and hippocampal subfields. Multiple linear regressions were conducted to test interactions between amyloid status and SDB severity on the volume of MTL subregions, controlling for age, sex, education, and the ApoE4 status. Secondary analyses aimed at investigating the links between SDB, MTL subregional atrophy, and episodic memory performance at baseline and at a mean follow-up of 20.66 months in the whole cohort and in subgroups stratified according to amyloid status. RESULTS We included 122 cognitively intact community-dwelling older adults (mean age ± SD: 69.40 ± 3.85 years, 77 women, 26 Aβ+ individuals) in baseline analyses and 111 at follow-up. The apnea-hypopnea index interacted with entorhinal (β = -0.81, p < 0.001, pη2 = 0.19), whole hippocampal (β = -0.61, p < 0.001, pη2 = 0.10), subiculum (β = -0.56, p = 0.002, pη2 = 0.08), CA1 (β = -0.55, p = 0.002, pη2 = 0.08), and DG (β = -0.53, p = 0.003, pη2 = 0.08) volumes such that a higher sleep apnea severity was related to lower MTL subregion volumes in amyloid-positive individuals, but not in those who were amyloid negative. In the whole cohort, lower whole hippocampal (r = 0.27, p = 0.005) and CA1 (r = 0.28, p = 0.003) volumes at baseline were associated with worse episodic memory performance at follow-up. DISCUSSION Overall, we showed that SDB was associated with MTL atrophy in cognitively asymptomatic older adults engaged in the Alzheimer continuum, which may increase the risk of developing memory impairment over time. TRIAL REGISTRATION INFORMATION ClinicalTrials.gov Identifier: NCT02977819.
Collapse
Affiliation(s)
- Claire André
- From the Normandie Univ (C.A., E.K., S.R., V.O., S.D.-S., C.P., F.F., P.C., S.D., D.V., G.C., R.F., G.R.), UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders" NEUROPRESAGE Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, France; Normandie Univ (C.A., S.R., V.O., P.C., V.L.S.), UNICAEN, PSL Université, EPHE, INSERM, CHU de Caen, GIP Cyceron, NIMH, France; Penn Image Computing and Science Laboratory (PICSL) (P.Y.), University of Pennsylvania, Philadelphia; Département de Recherche Clinique (D.V.), CHU Caen-Normandie, France; and Service de Neurologie (V.L.S.), CHU de Caen, France
| | - Elizabeth Kuhn
- From the Normandie Univ (C.A., E.K., S.R., V.O., S.D.-S., C.P., F.F., P.C., S.D., D.V., G.C., R.F., G.R.), UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders" NEUROPRESAGE Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, France; Normandie Univ (C.A., S.R., V.O., P.C., V.L.S.), UNICAEN, PSL Université, EPHE, INSERM, CHU de Caen, GIP Cyceron, NIMH, France; Penn Image Computing and Science Laboratory (PICSL) (P.Y.), University of Pennsylvania, Philadelphia; Département de Recherche Clinique (D.V.), CHU Caen-Normandie, France; and Service de Neurologie (V.L.S.), CHU de Caen, France
| | - Stéphane Rehel
- From the Normandie Univ (C.A., E.K., S.R., V.O., S.D.-S., C.P., F.F., P.C., S.D., D.V., G.C., R.F., G.R.), UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders" NEUROPRESAGE Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, France; Normandie Univ (C.A., S.R., V.O., P.C., V.L.S.), UNICAEN, PSL Université, EPHE, INSERM, CHU de Caen, GIP Cyceron, NIMH, France; Penn Image Computing and Science Laboratory (PICSL) (P.Y.), University of Pennsylvania, Philadelphia; Département de Recherche Clinique (D.V.), CHU Caen-Normandie, France; and Service de Neurologie (V.L.S.), CHU de Caen, France
| | - Valentin Ourry
- From the Normandie Univ (C.A., E.K., S.R., V.O., S.D.-S., C.P., F.F., P.C., S.D., D.V., G.C., R.F., G.R.), UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders" NEUROPRESAGE Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, France; Normandie Univ (C.A., S.R., V.O., P.C., V.L.S.), UNICAEN, PSL Université, EPHE, INSERM, CHU de Caen, GIP Cyceron, NIMH, France; Penn Image Computing and Science Laboratory (PICSL) (P.Y.), University of Pennsylvania, Philadelphia; Département de Recherche Clinique (D.V.), CHU Caen-Normandie, France; and Service de Neurologie (V.L.S.), CHU de Caen, France
| | - Solène Demeilliez-Servouin
- From the Normandie Univ (C.A., E.K., S.R., V.O., S.D.-S., C.P., F.F., P.C., S.D., D.V., G.C., R.F., G.R.), UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders" NEUROPRESAGE Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, France; Normandie Univ (C.A., S.R., V.O., P.C., V.L.S.), UNICAEN, PSL Université, EPHE, INSERM, CHU de Caen, GIP Cyceron, NIMH, France; Penn Image Computing and Science Laboratory (PICSL) (P.Y.), University of Pennsylvania, Philadelphia; Département de Recherche Clinique (D.V.), CHU Caen-Normandie, France; and Service de Neurologie (V.L.S.), CHU de Caen, France
| | - Cassandre Palix
- From the Normandie Univ (C.A., E.K., S.R., V.O., S.D.-S., C.P., F.F., P.C., S.D., D.V., G.C., R.F., G.R.), UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders" NEUROPRESAGE Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, France; Normandie Univ (C.A., S.R., V.O., P.C., V.L.S.), UNICAEN, PSL Université, EPHE, INSERM, CHU de Caen, GIP Cyceron, NIMH, France; Penn Image Computing and Science Laboratory (PICSL) (P.Y.), University of Pennsylvania, Philadelphia; Département de Recherche Clinique (D.V.), CHU Caen-Normandie, France; and Service de Neurologie (V.L.S.), CHU de Caen, France
| | - Francesca Felisatti
- From the Normandie Univ (C.A., E.K., S.R., V.O., S.D.-S., C.P., F.F., P.C., S.D., D.V., G.C., R.F., G.R.), UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders" NEUROPRESAGE Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, France; Normandie Univ (C.A., S.R., V.O., P.C., V.L.S.), UNICAEN, PSL Université, EPHE, INSERM, CHU de Caen, GIP Cyceron, NIMH, France; Penn Image Computing and Science Laboratory (PICSL) (P.Y.), University of Pennsylvania, Philadelphia; Département de Recherche Clinique (D.V.), CHU Caen-Normandie, France; and Service de Neurologie (V.L.S.), CHU de Caen, France
| | - Pierre Champetier
- From the Normandie Univ (C.A., E.K., S.R., V.O., S.D.-S., C.P., F.F., P.C., S.D., D.V., G.C., R.F., G.R.), UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders" NEUROPRESAGE Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, France; Normandie Univ (C.A., S.R., V.O., P.C., V.L.S.), UNICAEN, PSL Université, EPHE, INSERM, CHU de Caen, GIP Cyceron, NIMH, France; Penn Image Computing and Science Laboratory (PICSL) (P.Y.), University of Pennsylvania, Philadelphia; Département de Recherche Clinique (D.V.), CHU Caen-Normandie, France; and Service de Neurologie (V.L.S.), CHU de Caen, France
| | - Sophie Dautricourt
- From the Normandie Univ (C.A., E.K., S.R., V.O., S.D.-S., C.P., F.F., P.C., S.D., D.V., G.C., R.F., G.R.), UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders" NEUROPRESAGE Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, France; Normandie Univ (C.A., S.R., V.O., P.C., V.L.S.), UNICAEN, PSL Université, EPHE, INSERM, CHU de Caen, GIP Cyceron, NIMH, France; Penn Image Computing and Science Laboratory (PICSL) (P.Y.), University of Pennsylvania, Philadelphia; Département de Recherche Clinique (D.V.), CHU Caen-Normandie, France; and Service de Neurologie (V.L.S.), CHU de Caen, France
| | - Paul Yushkevich
- From the Normandie Univ (C.A., E.K., S.R., V.O., S.D.-S., C.P., F.F., P.C., S.D., D.V., G.C., R.F., G.R.), UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders" NEUROPRESAGE Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, France; Normandie Univ (C.A., S.R., V.O., P.C., V.L.S.), UNICAEN, PSL Université, EPHE, INSERM, CHU de Caen, GIP Cyceron, NIMH, France; Penn Image Computing and Science Laboratory (PICSL) (P.Y.), University of Pennsylvania, Philadelphia; Département de Recherche Clinique (D.V.), CHU Caen-Normandie, France; and Service de Neurologie (V.L.S.), CHU de Caen, France
| | - Denis Vivien
- From the Normandie Univ (C.A., E.K., S.R., V.O., S.D.-S., C.P., F.F., P.C., S.D., D.V., G.C., R.F., G.R.), UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders" NEUROPRESAGE Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, France; Normandie Univ (C.A., S.R., V.O., P.C., V.L.S.), UNICAEN, PSL Université, EPHE, INSERM, CHU de Caen, GIP Cyceron, NIMH, France; Penn Image Computing and Science Laboratory (PICSL) (P.Y.), University of Pennsylvania, Philadelphia; Département de Recherche Clinique (D.V.), CHU Caen-Normandie, France; and Service de Neurologie (V.L.S.), CHU de Caen, France
| | - Vincent de La Sayette
- From the Normandie Univ (C.A., E.K., S.R., V.O., S.D.-S., C.P., F.F., P.C., S.D., D.V., G.C., R.F., G.R.), UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders" NEUROPRESAGE Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, France; Normandie Univ (C.A., S.R., V.O., P.C., V.L.S.), UNICAEN, PSL Université, EPHE, INSERM, CHU de Caen, GIP Cyceron, NIMH, France; Penn Image Computing and Science Laboratory (PICSL) (P.Y.), University of Pennsylvania, Philadelphia; Département de Recherche Clinique (D.V.), CHU Caen-Normandie, France; and Service de Neurologie (V.L.S.), CHU de Caen, France
| | - Gaël Chételat
- From the Normandie Univ (C.A., E.K., S.R., V.O., S.D.-S., C.P., F.F., P.C., S.D., D.V., G.C., R.F., G.R.), UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders" NEUROPRESAGE Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, France; Normandie Univ (C.A., S.R., V.O., P.C., V.L.S.), UNICAEN, PSL Université, EPHE, INSERM, CHU de Caen, GIP Cyceron, NIMH, France; Penn Image Computing and Science Laboratory (PICSL) (P.Y.), University of Pennsylvania, Philadelphia; Département de Recherche Clinique (D.V.), CHU Caen-Normandie, France; and Service de Neurologie (V.L.S.), CHU de Caen, France
| | - Robin de Flores
- From the Normandie Univ (C.A., E.K., S.R., V.O., S.D.-S., C.P., F.F., P.C., S.D., D.V., G.C., R.F., G.R.), UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders" NEUROPRESAGE Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, France; Normandie Univ (C.A., S.R., V.O., P.C., V.L.S.), UNICAEN, PSL Université, EPHE, INSERM, CHU de Caen, GIP Cyceron, NIMH, France; Penn Image Computing and Science Laboratory (PICSL) (P.Y.), University of Pennsylvania, Philadelphia; Département de Recherche Clinique (D.V.), CHU Caen-Normandie, France; and Service de Neurologie (V.L.S.), CHU de Caen, France
| | - Géraldine Rauchs
- From the Normandie Univ (C.A., E.K., S.R., V.O., S.D.-S., C.P., F.F., P.C., S.D., D.V., G.C., R.F., G.R.), UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders" NEUROPRESAGE Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, France; Normandie Univ (C.A., S.R., V.O., P.C., V.L.S.), UNICAEN, PSL Université, EPHE, INSERM, CHU de Caen, GIP Cyceron, NIMH, France; Penn Image Computing and Science Laboratory (PICSL) (P.Y.), University of Pennsylvania, Philadelphia; Département de Recherche Clinique (D.V.), CHU Caen-Normandie, France; and Service de Neurologie (V.L.S.), CHU de Caen, France.
| |
Collapse
|
10
|
Arias-Cavieres A, Garcia AJ. A consequence of immature breathing induces persistent changes in hippocampal synaptic plasticity and behavior: a role of prooxidant state and NMDA receptor imbalance. Front Mol Neurosci 2023; 16:1192833. [PMID: 37456523 PMCID: PMC10338931 DOI: 10.3389/fnmol.2023.1192833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/24/2023] [Indexed: 07/18/2023] Open
Abstract
Underdeveloped breathing results from premature birth and causes intermittent hypoxia during the early neonatal period. Neonatal intermittent hypoxia (nIH) is a condition linked to the increased risk of neurocognitive deficit later in life. However, the mechanistic basis of nIH-induced changes to neurophysiology remains poorly resolved. We investigated the impact of nIH on hippocampal synaptic plasticity and NMDA receptor (NMDAr) expression in neonatal mice. Our findings indicate that nIH induces a prooxidant state that leads to an imbalance in NMDAr subunit composition favoring GluN2B over GluN2A expression and impairs synaptic plasticity. These consequences persist in adulthood and coincide with deficits in spatial memory. Treatment with an antioxidant, manganese (III) tetrakis (1-methyl-4-pyridyl)porphyrin (MnTMPyP), during nIH effectively mitigated both immediate and long-term effects of nIH. However, MnTMPyP treatment post-nIH did not prevent long-lasting changes in either synaptic plasticity or behavior. In addition to demonstrating that the prooxidant state has a central role in nIH-mediated neurophysiological and behavioral deficits, our results also indicate that targeting the prooxidant state during a discrete therapeutic window may provide a potential avenue for mitigating long-term neurophysiological and behavioral outcomes that result from unstable breathing during early postnatal life.
Collapse
Affiliation(s)
- Alejandra Arias-Cavieres
- Institute for Integrative Physiology, The University of Chicago, Chicago, IL, United States
- Department of Medicine, Section of Emergency Medicine, The University of Chicago, Chicago, IL, United States
| | - Alfredo J. Garcia
- Institute for Integrative Physiology, The University of Chicago, Chicago, IL, United States
- Department of Medicine, Section of Emergency Medicine, The University of Chicago, Chicago, IL, United States
- University of Chicago Neuroscience Institute, University of Chicago, Chicago, IL, United States
| |
Collapse
|
11
|
Zhang Y, Miao Y, Xiong X, Tan J, Han Z, Chen F, Lei P, Zhang Q. Microglial exosomes alleviate intermittent hypoxia-induced cognitive deficits by suppressing NLRP3 inflammasome. Biol Direct 2023; 18:29. [PMID: 37312196 DOI: 10.1186/s13062-023-00387-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 06/01/2023] [Indexed: 06/15/2023] Open
Abstract
Intermittent hypoxia is the best predictor of developing cognitive decline and Alzheimer's disease progression in patients with obstructive sleep apnea. The nucleotide-binding oligomerization domain-like receptor 3 (NLRP3) inflammasome has been poorly studied as a regulator of neuroinflammation in cognitive impairment caused by intermittent hypoxia. As critical inflammatory cells, exosomes secreted by microglia have been found to affect the spread of pathologic proteins and neuropathology in neurodegenerative diseases. However, the effects of microglial exosomes on neuroinflammation and cognitive outcomes after intermittent hypoxia remain unclear. In this study, the role of miRNAs in microglial exosomes in improving cognitive deficits in mice exposed to intermittent hypoxia was investigated. We demonstrated that miR-146a-5p fluctuated over time in microglial exosomes of mice exposed to intermittent hypoxia for different periods of time, which could regulate neuronal NLRP3 inflammasome and neuroinflammation. In primary neurons, we found that miR-146a-5p regulated mitochondrial reactive oxygen species by targeting HIF1α, thus affecting the NLRP3 inflammasome and secretion of inflammatory factors. Similarly, further studies showed that inhibition of NLRP3 by administering overexpressed miR-146a-5p in microglial exosomes and MCC950 has improved neuroinflammation and cognitive dysfunction in mice after intermittent hypoxia. In conclusion, NLRP3 inflammasome may be a regulatory target for ameliorating cognitive impairment caused by intermittent hypoxia, and microglial exosomal miR-146a-5p may be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yaodan Zhang
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin, 300052, China
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yuyang Miao
- Tianjin Medical University, Tianjin, 300052, China
| | - Xiangyang Xiong
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin, 300052, China
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jin Tan
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin, 300052, China
| | - Zhaoli Han
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin, 300052, China
| | - Fanglian Chen
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Ping Lei
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin, 300052, China.
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Qiang Zhang
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin, 300052, China.
| |
Collapse
|
12
|
Ramirez JM, Carroll MS, Burgraff N, Rand CM, Weese-Mayer DE. A narrative review of the mechanisms and consequences of intermittent hypoxia and the role of advanced analytic techniques in pediatric autonomic disorders. Clin Auton Res 2023; 33:287-300. [PMID: 37326924 DOI: 10.1007/s10286-023-00958-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 05/25/2023] [Indexed: 06/17/2023]
Abstract
Disorders of autonomic functions are typically characterized by disturbances in multiple organ systems. These disturbances are often comorbidities of common and rare diseases, such as epilepsy, sleep apnea, Rett syndrome, congenital heart disease or mitochondrial diseases. Characteristic of many autonomic disorders is the association with intermittent hypoxia and oxidative stress, which can cause or exaggerate a variety of other autonomic dysfunctions, making the treatment and management of these syndromes very complex. In this review we discuss the cellular mechanisms by which intermittent hypoxia can trigger a cascade of molecular, cellular and network events that result in the dysregulation of multiple organ systems. We also describe the importance of computational approaches, artificial intelligence and the analysis of big data to better characterize and recognize the interconnectedness of the various autonomic and non-autonomic symptoms. These techniques can lead to a better understanding of the progression of autonomic disorders, ultimately resulting in better care and management.
Collapse
Affiliation(s)
- Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA, 98101, USA.
- Departments of Neurological Surgery and Pediatrics, University of Washington School of Medicine, 1900 Ninth Avenue, Seattle, WA, 98101, USA.
| | - Michael S Carroll
- Data Analytics and Reporting, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Autonomic Medicine, Stanley Manne Children's Research Institute at Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Nicholas Burgraff
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA, 98101, USA
| | - Casey M Rand
- Division of Autonomic Medicine, Stanley Manne Children's Research Institute at Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Debra E Weese-Mayer
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Autonomic Medicine, Stanley Manne Children's Research Institute at Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| |
Collapse
|
13
|
Puzio M, Moreton N, Sullivan M, Scaife C, Glennon JC, O'Connor JJ. An Electrophysiological and Proteomic Analysis of the Effects of the Superoxide Dismutase Mimetic, MnTMPyP, on Synaptic Signalling Post-Ischemia in Isolated Rat Hippocampal Slices. Antioxidants (Basel) 2023; 12:antiox12040792. [PMID: 37107167 PMCID: PMC10135248 DOI: 10.3390/antiox12040792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Metabolic stress and the increased production of reactive oxygen species (ROS) are two main contributors to neuronal damage and synaptic plasticity in acute ischemic stroke. The superoxide scavenger MnTMPyP has been previously reported to have a neuroprotective effect in organotypic hippocampal slices and to modulate synaptic transmission after in vitro hypoxia and oxygen-glucose deprivation (OGD). However, the mechanisms involved in the effect of this scavenger remain elusive. In this study, two concentrations of MnTMPyP were evaluated on synaptic transmission during ischemia and post-ischemic synaptic potentiation. The complex molecular changes supporting cellular adaptation to metabolic stress, and how these are modulated by MnTMPyP, were also investigated. Electrophysiological data showed that MnTMPyP causes a decrease in baseline synaptic transmission and impairment of synaptic potentiation. Proteomic analysis performed on MnTMPyP and hypoxia-treated tissue indicated an impairment in vesicular trafficking mechanisms, including reduced expression of Hsp90 and actin signalling. Alterations of vesicular trafficking may lead to reduced probability of neurotransmitter release and AMPA receptor activity, resulting in the observed modulatory effect of MnTMPyP. In OGD, protein enrichment analysis highlighted impairments in cell proliferation and differentiation, such as TGFβ1 and CDKN1B signalling, in addition to downregulation of mitochondrial dysfunction and an increased expression of CAMKII. Taken together, our results may indicate modulation of neuronal sensitivity to the ischemic insult, and a complex role for MnTMPyP in synaptic transmission and plasticity, potentially providing molecular insights into the mechanisms mediating the effects of MnTMPyP during ischemia.
Collapse
Affiliation(s)
- Martina Puzio
- UCD School of Biomolecular & Biomedical Science, University College Dublin, Dublin 4, Ireland
- Mass Spectrometry Core Facility, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Niamh Moreton
- UCD School of Biomolecular & Biomedical Science, University College Dublin, Dublin 4, Ireland
- Mass Spectrometry Core Facility, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Mairéad Sullivan
- Mass Spectrometry Core Facility, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
- UCD School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Caitriona Scaife
- Mass Spectrometry Core Facility, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Jeffrey C Glennon
- Mass Spectrometry Core Facility, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
- UCD School of Medicine, University College Dublin, Dublin 4, Ireland
| | - John J O'Connor
- UCD School of Biomolecular & Biomedical Science, University College Dublin, Dublin 4, Ireland
- Mass Spectrometry Core Facility, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| |
Collapse
|
14
|
Arias-Cavieres A, Garcia AJ. A Consequence of Immature Breathing induces Persistent Changes in Hippocampal Synaptic Plasticity and Behavior: A Role of Pro-Oxidant State and NMDA Receptor Imbalance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.21.533692. [PMID: 36993632 PMCID: PMC10055328 DOI: 10.1101/2023.03.21.533692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Underdeveloped breathing results from premature birth and causes intermittent hypoxia during the early neonatal period. Neonatal intermittent hypoxia (nIH) is a condition linked to the increased risk of neurocognitive deficit later in life. However, the underlying mechanistic consequences nIH-induced neurophysiological changes remains poorly resolved. Here, we investigated the impact of nIH on hippocampal synaptic plasticity and NMDA receptor (NMDAr) expression in neonatal mice. Our findings indicate that nIH induces a pro-oxidant state, leading to an imbalance in NMDAr subunit composition that favors GluN2A over GluN2B expression, and subsequently impairs synaptic plasticity. These consequences persist in adulthood and coincide with deficits in spatial memory. Treatment with the antioxidant, manganese(III) tetrakis(1-methyl-4-pyridyl)porphyrin (MnTMPyP), during nIH effectively mitigated both immediate and long-term effects of nIH. However, MnTMPyP treatment post-nIH did not prevent the long-lasting changes in either synaptic plasticity or behavior. Our results underscore the central role of the pro-oxidant state in nIH-mediated neurophysiological and behavioral deficits and importance of stable oxygen homeostasis during early life. These findings suggest that targeting the pro-oxidant state during a discrete window may provide a potential avenue for mitigating long-term neurophysiological and behavioral outcomes when breathing is unstable during early postnatal life. Highlights Untreated immature breathing leads neonatal intermittent hypoxia (nIH).nIH promotes a pro-oxidant state associated with increased HIF1a activity and NOX upregulation.nIH-dependent pro-oxidant state leads to NMDAr remodeling of the GluN2 subunit to impair synaptic plasticity.Impaired synaptic plasticity and NMDAr remodeling caused by nIH persists beyond the critical period of development.A discrete window for antioxidant administration exists to effectively mitigate neurophysiological and behavioral consequences of nIH.
Collapse
Affiliation(s)
- Alejandra Arias-Cavieres
- Institute for Integrative Physiology, The University of Chicago
- Department of Medicine, Section of Emergency Medicine, The University of Chicago
| | - Alfredo J. Garcia
- Institute for Integrative Physiology, The University of Chicago
- Grossman Institute for Neuroscience, Quantitative Biology & Human Behavior, The University of Chicago
- Department of Medicine, Section of Emergency Medicine, The University of Chicago
| |
Collapse
|
15
|
Yang XY, Geng L, Li R, Song JX, Jia CL, An JR, Sun MF, Xu S, Guo YJ, Zhao Y, Ji ES. Huperzine A-Liposomes Efficiently Improve Neural Injury in the Hippocampus of Mice with Chronic Intermittent Hypoxia. Int J Nanomedicine 2023; 18:843-859. [PMID: 36824413 PMCID: PMC9942512 DOI: 10.2147/ijn.s393346] [Citation(s) in RCA: 3] [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: 10/22/2022] [Accepted: 02/07/2023] [Indexed: 02/18/2023] Open
Abstract
Background Chronic intermittent hypoxia (CIH) could cause neuronal damage, accelerating the progression of dementia. However, safe and effective therapeutic drugs and delivery are needed for successful CIH therapy. Purpose To investigate the neuroprotective effect of Huperzine A (HuA) packaged with nanoliposomes (HuA-LIP) on neuronal damage induced by CIH. Methods The stability and release of HuA-LIP in vitro were identified. Mice were randomly divided into the Control, CIH, HuA-LIP, and HuA groups. The mice in the HuA and HuA-LIP groups received HuA (0.1 mg/kg, i.p.), and HuA-LIP was administered during CIH exposure for 21 days. HuA-LIP contains the equivalent content of HuA. Results We prepared a novel formulation of HuA-LIP that had good stability and controlled release. First, HuA-LIP significantly ameliorated cognitive dysfunction and neuronal damage in CIH mice. Second, HuA-LIP elevated T-SOD and GSH-Px abilities and decreased MDA content to resist oxidative stress damage induced by CIH. Furthermore, HuA-LIP reduced brain iron levels by downregulating TfR1, hepcidin, and FTL expression. In addition, HuA-LIP activated the PKAα/Erk/CREB/BDNF signaling pathway and elevated MAP2, PSD95, and synaptophysin to improve synaptic plasticity. Most importantly, compared with HuA, HuA-LIP showed a superior performance against neuronal damage induced by CIH. Conclusion HuA-LIP has a good sustained-release effect and targeting ability and efficiently protects against neural injury caused by CIH.
Collapse
Affiliation(s)
- Xin-Yue Yang
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, People’s Republic of China
| | - Lina Geng
- College of Chemistry and Material Science, Hebei Normal University, Shijiazhuang, People’s Republic of China
| | - Ronghui Li
- College of Chemistry and Material Science, Hebei Normal University, Shijiazhuang, People’s Republic of China
| | - Ji-Xian Song
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, People’s Republic of China
| | - Cui-Ling Jia
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, People’s Republic of China
| | - Ji-Ren An
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, People’s Republic of China
- The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, People’s Republic of China
| | - Meng-Fan Sun
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, People’s Republic of China
| | - Shan Xu
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, People’s Republic of China
| | - Ya-Jing Guo
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, People’s Republic of China
| | - Yashuo Zhao
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, People’s Republic of China
| | - En-Sheng Ji
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, People’s Republic of China
| |
Collapse
|
16
|
Li G, Guan Y, Gu Y, Guo M, Ma W, Shao Q, Liu J, Ji X. Intermittent hypoxic conditioning restores neurological dysfunction of mice induced by long-term hypoxia. CNS Neurosci Ther 2022; 29:202-215. [PMID: 36401601 PMCID: PMC9804074 DOI: 10.1111/cns.13996] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 09/18/2022] [Accepted: 09/20/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Central nervous system diseases are associated with hypoxia, which usually cause irreversible nerve damage, but the underlying mechanism is unclear and effective intervention strategies are lacking. This study was designed to explore the mechanism and treatment strategy of hypoxia-induced nerve injury. METHODS In this study, 13% O2 was used to treat mice for 0, 1, 3 7, and 14 days, Morris water maze and other animal behavior experiments were used to evaluate the neurological function of mice. TUNEL, BrdU, PCNA, DCX, and SOX2 staining were used to observe the apoptosis and proliferation of mouse neurons. RT-PCR and Iba1 staining were used to evaluate the release of inflammatory factors IL-1β, IL-6, and TNF-α and the activation of microglia. RESULTS Short-term hypoxia promotes neurogenesis, while long-term hypoxia inhibits neurogenesis. The changes in hypoxia-induced neurogenesis were positively correlated with neurological functions, but negatively correlated with apoptosis. Moreover, intermittent hypoxic conditioning restored long-term hypoxia-induced neurological dysfunction by promoting neural stem cell generation and inhibiting the release of inflammatory factors IL-1β, IL-6, and TNF-α and the activation of microglia. CONCLUSION Hypoxia promoted neurogenesis in a time-dependent manner, and intermittent hypoxic conditioning exerted a neuroprotective effect through promoting neural stem cell generation and suppressing inflammation induced by long-term hypoxia stress, which provided a novel concept to develop a treatment for hypoxia-related brain injury.
Collapse
Affiliation(s)
- Gaifen Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina,Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Yuying Guan
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina,Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Yakun Gu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Mengyuan Guo
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Wei Ma
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Qianqian Shao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Jia Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina,Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
17
|
Puech C, Badran M, Runion AR, Barrow MB, Qiao Z, Khalyfa A, Gozal D. Explicit memory, anxiety and depressive like behavior in mice exposed to chronic intermittent hypoxia, sleep fragmentation, or both during the daylight period. Neurobiol Sleep Circadian Rhythms 2022; 13:100084. [PMID: 36254342 PMCID: PMC9568859 DOI: 10.1016/j.nbscr.2022.100084] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/01/2022] [Accepted: 10/07/2022] [Indexed: 11/27/2022] Open
Abstract
Obstructive sleep apnea (OSA) is a chronic and highly prevalent condition characterized by chronic intermittent hypoxia (IH) and sleep fragmentation (SF), and can lead to a vast array of end-organ morbidities, particularly affecting cardiovascular, metabolic and neurobehavioral functioning. OSA can induce cognitive and behavioral and mood deficits. Male C57Bl/6J 8-week-old mice were housed in custom-designed cages with a silent motorized mechanical sweeper traversing the cage floor at 2-min intervals (SF) during daylight for four weeks. Sleep control (SC) consisted of keeping sweeper immobile. IH consisted of cycling FiO2 21% 90 seconds-6.3% 90s or room air (RA; FiO2 21%) for sixteen weeks and combined SF-IH was conducted for nine weeks. Open field novel object recognition (NOR) testing, elevated-plus maze test (EPMT), and forced swimming test (FST) were performed. SF induced cognitive NOR performance impairments in mice along with reduced anxiety behaviors while IH induced deficits in NOR performance, but increased anxiety behaviors. SF-IH induced impaired performance in NOR test of similar magnitude to IH or SF alone. Combined SF-IH exposures did not affect anxiety behaviors. Thus, both SF an IH altered cognitive function while imposing opposite effects on anxiety behaviors. SF-IH did not magnify the detrimental effects of isolated SF or IH and canceled out the effects on anxiety. Based on these findings, the underlying pathophysiologic processes underlying IH and SF adverse effects on cognitive function appear to differ, while those affecting anxiety counteract each other.
Collapse
Affiliation(s)
- Clementine Puech
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Mohammad Badran
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Alexandra R Runion
- Undergraduate Student Research Program, University of Missouri, Columbia, MO, USA
| | - Max B Barrow
- Undergraduate Student Research Program, University of Missouri, Columbia, MO, USA
| | - Zhuanhong Qiao
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Abdelnaby Khalyfa
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - David Gozal
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
18
|
Rangan P, Lobo F, Parrella E, Rochette N, Morselli M, Stephen TL, Cremonini AL, Tagliafico L, Persia A, Caffa I, Monacelli F, Odetti P, Bonfiglio T, Nencioni A, Pigliautile M, Boccardi V, Mecocci P, Pike CJ, Cohen P, LaDu MJ, Pellegrini M, Xia K, Tran K, Ann B, Chowdhury D, Longo VD. Fasting-mimicking diet cycles reduce neuroinflammation to attenuate cognitive decline in Alzheimer's models. Cell Rep 2022; 40:111417. [PMID: 36170815 PMCID: PMC9648488 DOI: 10.1016/j.celrep.2022.111417] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/30/2022] [Accepted: 09/01/2022] [Indexed: 11/22/2022] Open
Abstract
The effects of fasting-mimicking diet (FMD) cycles in reducing many aging and disease risk factors indicate it could affect Alzheimer's disease (AD). Here, we show that FMD cycles reduce cognitive decline and AD pathology in E4FAD and 3xTg AD mouse models, with effects superior to those caused by protein restriction cycles. In 3xTg mice, long-term FMD cycles reduce hippocampal Aβ load and hyperphosphorylated tau, enhance genesis of neural stem cells, decrease microglia number, and reduce expression of neuroinflammatory genes, including superoxide-generating NADPH oxidase (Nox2). 3xTg mice lacking Nox2 or mice treated with the NADPH oxidase inhibitor apocynin also display improved cognition and reduced microglia activation compared with controls. Clinical data indicate that FMD cycles are feasible and generally safe in a small group of AD patients. These results indicate that FMD cycles delay cognitive decline in AD models in part by reducing neuroinflammation and/or superoxide production in the brain.
Collapse
Affiliation(s)
- Priya Rangan
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Fleur Lobo
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Edoardo Parrella
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA; Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, BS 25123, Italy
| | - Nicolas Rochette
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, 611 Charles E. Young Dr. E., Los Angeles, CA 90095, USA; Department of Ecology and Evolutionary Biology, University of California, Los Angeles, 612 Charles E. Young Dr. E., Los Angeles, CA 90095, USA
| | - Marco Morselli
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 610 Charles E. Young Dr. S., Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, 611 Charles E. Young Dr. E., Los Angeles, CA 90095, USA
| | - Terri-Leigh Stephen
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Anna Laura Cremonini
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Luca Tagliafico
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy
| | - Angelica Persia
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy
| | - Irene Caffa
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy
| | - Fiammetta Monacelli
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Patrizio Odetti
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Tommaso Bonfiglio
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Martina Pigliautile
- Santa Maria della Misericordia Hospital, Section of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Virginia Boccardi
- Santa Maria della Misericordia Hospital, Section of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Patrizia Mecocci
- Santa Maria della Misericordia Hospital, Section of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Christian J Pike
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Pinchas Cohen
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA; USC Dornsife College of Letters, Arts & Sciences, Department of Biological Sciences, University of Southern California, 3551 Trousdale Pkwy., Los Angeles, CA 90089-0191, USA
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 610 Charles E. Young Dr. S., Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, 611 Charles E. Young Dr. E., Los Angeles, CA 90095, USA
| | - Kyle Xia
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Katelynn Tran
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Brandon Ann
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Dolly Chowdhury
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Valter D Longo
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA; Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, 1425 San Pablo St., Los Angeles, CA 90033, USA; IFOM FIRC Institute of Molecular Oncology, Via Adamello 16, Milano, MI 20139, Italy.
| |
Collapse
|
19
|
Inhibition of STAT3 signal pathway recovers postsynaptic plasticity to improve cognitive impairment caused by chronic intermittent hypoxia. Sleep Breath 2022; 27:893-902. [DOI: 10.1007/s11325-022-02671-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 06/08/2022] [Accepted: 06/16/2022] [Indexed: 10/16/2022]
|
20
|
Neuroprotective strategies for acute ischemic stroke: Targeting oxidative stress and prolyl hydroxylase domain inhibition in synaptic signalling. BRAIN DISORDERS 2022. [DOI: 10.1016/j.dscb.2022.100030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
21
|
Chronic Intermittent Hypoxia Enhances Pathological Tau Seeding, Propagation, and Accumulation and Exacerbates Alzheimer-like Memory and Synaptic Plasticity Deficits and Molecular Signatures. Biol Psychiatry 2022; 91:346-358. [PMID: 34130857 PMCID: PMC8895475 DOI: 10.1016/j.biopsych.2021.02.973] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 02/19/2021] [Accepted: 02/28/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Obstructive sleep apnea, characterized by sleep fragmentation and chronic intermittent hypoxia (CIH), is a risk factor for Alzheimer's disease (AD) progression. Recent epidemiological studies point to CIH as the best predictor of developing cognitive decline and AD in older adults with obstructive sleep apnea. However, the precise underlying mechanisms remain unknown. This study was undertaken to evaluate the effect of CIH on pathological human tau seeding, propagation, and accumulation; cognition; synaptic plasticity; neuronal network excitability; and gene expression profiles in a P301S human mutant tau mouse model of AD and related tauopathies. METHODS We exposed 4- to 4.5-month-old male P301S and wild-type mice to an 8-week CIH protocol (6-min cycle: 21% O2 to 8% O2 to 21% O2, 80 cycles per 8 hours during daytime) and assessed its effect on tau pathology and various AD-related phenotypic and molecular signatures. Age- and sex-matched P301S and wild-type mice were reared in normoxia (21% O2) as experimental controls. RESULTS CIH significantly enhanced pathological human tau seeding and spread across connected brain circuitry in P301S mice; it also increased phosphorylated tau load. CIH also exacerbated memory and synaptic plasticity deficits in P301S mice. However, CIH had no effect on seizure susceptibility and network hyperexcitability in these mice. Finally, CIH exacerbated AD-related pathogenic molecular signaling in P301S mice. CONCLUSIONS CIH-induced increase in pathologic human tau seeding and spread and exacerbation of other AD-related impairments provide new insights into the role of CIH and obstructive sleep apnea in AD pathogenesis.
Collapse
|
22
|
Ousta A, Piao L, Fang YH, Vera A, Nallamothu T, Garcia AJ, Sharp WW. Microglial Activation and Neurological Outcomes in a Murine Model of Cardiac Arrest. Neurocrit Care 2022; 36:61-70. [PMID: 34268646 PMCID: PMC8813848 DOI: 10.1007/s12028-021-01253-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 04/08/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Neurological injury following successful resuscitation from sudden cardiac arrest (CA) is common. The pathophysiological basis of this injury remains poorly understood, and treatment options are limited. Microglial activation and neuroinflammation are established contributors to many neuropathologies, such as Alzheimer disease and traumatic brain injury, but their potential role in post-CA injury has only recently been recognized. Here, we hypothesize that microglial activation that occurs following brief asystolic CA is associated with neurological injury and represents a potential therapeutic target. METHODS Adult C57BL/6 male and female mice were randomly assigned to 12-min, KCl-induced asystolic CA, under anesthesia and ventilation, followed by successful cardiopulmonary resuscitation (n = 19) or sham intervention (n = 11). Neurological assessments of mice were performed using standardized neurological scoring, video motion tracking, and sensory/motor testing. Mice were killed at 72 h for histological studies; neuronal degeneration was assessed using Fluoro-Jade C staining. Microglial characteristics were assessed by immunohistochemistry using the marker of ionized calcium binding adaptor molecule 1, followed by ImageJ analyses for cell integrity density and skeletal analyses. RESULTS Neurological injury in post-cardiopulmonary-resuscitation mice vs. sham mice was evident by poorer neurological scores (difference of 3.626 ± 0.4921, 95% confidence interval 2.618-4.634), sensory and motor functions (worsened by sixfold and sevenfold, respectively, compared with baseline), and locomotion (75% slower with a 76% decrease in total distance traveled). Post-CA brains demonstrated evidence of neurodegeneration and neuroinflammatory microglial activation. CONCLUSIONS Extensive microglial activation and neurodegeneration in the CA1 region and the dentate gyrus of the hippocampus are evident following brief asystolic CA and are associated with severe neurological injury.
Collapse
Affiliation(s)
- Alaa Ousta
- Section of Emergency Medicine, Department of Medicine, University of Chicago, 5841 S Maryland Avenue, Chicago, IL, 60637, USA
| | - Lin Piao
- Section of Emergency Medicine, Department of Medicine, University of Chicago, 5841 S Maryland Avenue, Chicago, IL, 60637, USA
| | - Yong Hu Fang
- Section of Emergency Medicine, Department of Medicine, University of Chicago, 5841 S Maryland Avenue, Chicago, IL, 60637, USA
| | - Adrianna Vera
- Section of Emergency Medicine, Department of Medicine, University of Chicago, 5841 S Maryland Avenue, Chicago, IL, 60637, USA
| | - Thara Nallamothu
- Section of Emergency Medicine, Department of Medicine, University of Chicago, 5841 S Maryland Avenue, Chicago, IL, 60637, USA
| | - Alfredo J Garcia
- Section of Emergency Medicine, Department of Medicine, University of Chicago, 5841 S Maryland Avenue, Chicago, IL, 60637, USA
| | - Willard W Sharp
- Section of Emergency Medicine, Department of Medicine, University of Chicago, 5841 S Maryland Avenue, Chicago, IL, 60637, USA.
| |
Collapse
|
23
|
Chauhan G, Kumar G, Roy K, Kumari P, Thondala B, Kishore K, Panjwani U, Ray K. Hypobaric Hypoxia Induces Deficits in Adult Neurogenesis and Social Interaction via Cyclooxygenase-1/ EP1 Receptor Pathway Activating NLRP3 Inflammasome. Mol Neurobiol 2022; 59:2497-2519. [PMID: 35089581 DOI: 10.1007/s12035-022-02750-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 01/11/2022] [Indexed: 12/17/2022]
Abstract
Low oxygen environments, like hypobaric hypoxia (HH), are common nodes in a number of diseases characterized by neuroinflammation, which is detrimental to the structural and functional aspects of hippocampal circuitry. Hypoxic conditions lead to elevation of inflammasome-mediated inflammation that may contribute to cognitive deficits. However, a systematic investigation of the impact of inflammasome-mediated neuroinflammation on the components of neurogenic niche during HH remains to be elusive. Cerebral hypoxia was induced in adult male Sprague Dawley rats via decreasing partial pressure of oxygen. The effect of HH (1, 3, and 7 days at 25,000 ft) on social memory, anxiety, adult neurogenesis, and NLRP3- (NLR family pyrin domain containing 3) mediated neuroinflammation in the dentate gyrus (DG) was explored in detail. Furthermore, we explored the therapeutic efficacy of cyclooxygenase-1 inhibitor (valeryl salicylate, 5 mg/kg/day, i.p.) and EP1 receptor (EP1R) antagonist (SC19220, 1 mg/kg/day, i.p.) on HH-induced deficits. Seven days of HH exposure induced alteration in social and anxiety-like behavior along with perturbation in adult neurogenesis. Elevation in NLRP3, caspase-1, and IL-1β levels was observed during HH from day 1. A notable increase in the COX-1/EP1R pathway in activated glial cells in DG was evident during HH. COX-1 inhibitor and EP1R antagonist mitigated the detrimental effects of HH on social memory, adult neurogenesis via blunting NLRP3-mediated inflammation. Our data showed induction of the COX-1/EP1R pathway in the glial cells, which is detrimental to neurogenesis and social memory, opening up the possibility that the COX-1/EP1R pathway is a plausible target for inflammasome-related neurogenesis impairments.
Collapse
Affiliation(s)
- Garima Chauhan
- Neurophysiology Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Gaurav Kumar
- Neurophysiology Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Koustav Roy
- Neurophysiology Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Punita Kumari
- Neurophysiology Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Bhanuteja Thondala
- Neurophysiology Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Krishna Kishore
- Neurophysiology Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Usha Panjwani
- Neurophysiology Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Koushik Ray
- Neurophysiology Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
24
|
Filchenko I, Korostovtseva L, Bochkarev M, Sviryaev Y. Brain damage in sleep-disordered breathing: the role of glia. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:15-22. [DOI: 10.17116/jnevro202212201115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
25
|
Kravtsov A, Kozin S, Basov A, Butina E, Baryshev M, Malyshko V, Moiseev A, Elkina A, Dzhimak S. Reduction of Deuterium Level Supports Resistance of Neurons to Glucose Deprivation and Hypoxia: Study in Cultures of Neurons and on Animals. Molecules 2021; 27:243. [PMID: 35011474 PMCID: PMC8746303 DOI: 10.3390/molecules27010243] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/29/2021] [Accepted: 12/29/2021] [Indexed: 11/17/2022] Open
Abstract
The effect of a reduced deuterium (D) content in the incubation medium on the survival of cultured neurons in vitro and under glucose deprivation was studied. In addition, we studied the effect of a decrease in the deuterium content in the rat brain on oxidative processes in the nervous tissue, its antioxidant protection, and training of rats in the T-shaped maze test under hypoxic conditions. For experiments with cultures of neurons, 7-8-day cultures of cerebellar neurons were used. Determination of the rate of neuronal death in cultures was carried out using propidium iodide. Acute hypoxia with hypercapnia was simulated in rats by placing them in sealed vessels with a capacity of 1 L. The effect on oxidative processes in brain tissues was assessed by changes in the level of free radical oxidation and malondialdehyde. The effect on the antioxidant system of the brain was assessed by the activity of catalase. The study in the T-maze was carried out in accordance with the generally accepted methodology, the skill of alternating right-sided and left-sided loops on positive reinforcement was developed. This work has shown that a decrease in the deuterium content in the incubation medium to a level of -357‱ has a neuroprotective effect, increasing the survival rate of cultured neurons under glucose deprivation. When exposed to hypoxia, a preliminary decrease in the deuterium content in the rat brain to -261‱ prevents the development of oxidative stress in their nervous tissue and preserves the learning ability of animals in the T-shaped maze test at the level of the control group. A similar protective effect during the modification of the 2H/1H internal environment of the body by the consumption of DDW can potentially be used for the prevention of pathological conditions associated with the development of oxidative stress with damage to the central nervous system.
Collapse
Affiliation(s)
- Alexandr Kravtsov
- Department of Radiophysics and Nanothechnology, Physics Faculty, Kuban State University, 350040 Krasnodar, Russia; (A.K.); (S.K.); (A.B.); (M.B.); (A.E.)
- South Scientific Center of the Russian Academy of Sciences, Laboratory of Problems of Stable Isotope Spreading in Living Systems, 344006 Rostov-on-Don, Russia;
| | - Stanislav Kozin
- Department of Radiophysics and Nanothechnology, Physics Faculty, Kuban State University, 350040 Krasnodar, Russia; (A.K.); (S.K.); (A.B.); (M.B.); (A.E.)
- South Scientific Center of the Russian Academy of Sciences, Laboratory of Problems of Stable Isotope Spreading in Living Systems, 344006 Rostov-on-Don, Russia;
| | - Alexandr Basov
- Department of Radiophysics and Nanothechnology, Physics Faculty, Kuban State University, 350040 Krasnodar, Russia; (A.K.); (S.K.); (A.B.); (M.B.); (A.E.)
- Department of Fundamental and Clinical Biochemistry, Kuban State Medical University, 350063 Krasnodar, Russia
| | - Elena Butina
- Department of Technology of Fats, Cosmetics, Commodity Science, Processes and Devices, Kuban State Technological University, 350072 Krasnodar, Russia;
| | - Mikhail Baryshev
- Department of Radiophysics and Nanothechnology, Physics Faculty, Kuban State University, 350040 Krasnodar, Russia; (A.K.); (S.K.); (A.B.); (M.B.); (A.E.)
- South Scientific Center of the Russian Academy of Sciences, Laboratory of Problems of Stable Isotope Spreading in Living Systems, 344006 Rostov-on-Don, Russia;
- Department of Technology of Fats, Cosmetics, Commodity Science, Processes and Devices, Kuban State Technological University, 350072 Krasnodar, Russia;
| | - Vadim Malyshko
- South Scientific Center of the Russian Academy of Sciences, Laboratory of Problems of Stable Isotope Spreading in Living Systems, 344006 Rostov-on-Don, Russia;
- Department of Fundamental and Clinical Biochemistry, Kuban State Medical University, 350063 Krasnodar, Russia
| | - Arkady Moiseev
- Department of Organization and Support of Scientific Activities, Kuban State Agrarian University, 350044 Krasnodar, Russia;
| | - Anna Elkina
- Department of Radiophysics and Nanothechnology, Physics Faculty, Kuban State University, 350040 Krasnodar, Russia; (A.K.); (S.K.); (A.B.); (M.B.); (A.E.)
- South Scientific Center of the Russian Academy of Sciences, Laboratory of Problems of Stable Isotope Spreading in Living Systems, 344006 Rostov-on-Don, Russia;
- Department of Physics, K.G. Razumovsky Moscow State University of Technologies and Management (The First Cossack University), 109004 Moscow, Russia
| | - Stepan Dzhimak
- Department of Radiophysics and Nanothechnology, Physics Faculty, Kuban State University, 350040 Krasnodar, Russia; (A.K.); (S.K.); (A.B.); (M.B.); (A.E.)
- South Scientific Center of the Russian Academy of Sciences, Laboratory of Problems of Stable Isotope Spreading in Living Systems, 344006 Rostov-on-Don, Russia;
- The V.M. Gorbatov Federal Research Center for Food Systems of the Russian Academy of Sciences, Experimental Clinic—Laboratory of Biologically Active Substances of Animal Origin, 109316 Moscow, Russia
| |
Collapse
|
26
|
Li G, Liu J, Guan Y, Ji X. The role of hypoxia in stem cell regulation of the central nervous system: From embryonic development to adult proliferation. CNS Neurosci Ther 2021; 27:1446-1457. [PMID: 34817133 PMCID: PMC8611781 DOI: 10.1111/cns.13754] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/28/2021] [Accepted: 10/03/2021] [Indexed: 12/12/2022] Open
Abstract
Hypoxia is involved in the regulation of various cell functions in the body, including the regulation of stem cells. The hypoxic microenvironment is indispensable from embryonic development to the regeneration and repair of adult cells. In addition to embryonic stem cells, which need to maintain their self-renewal properties and pluripotency in a hypoxic environment, adult stem cells, including neural stem cells (NSCs), also exist in a hypoxic microenvironment. The subventricular zone (SVZ) and hippocampal dentate gyrus (DG) are the main sites of adult neurogenesis in the brain. Hypoxia can promote the proliferation, migration, and maturation of NSCs in these regions. Also, because most neurons in the brain are non-regenerative, stem cell transplantation is considered as a promising strategy for treating central nervous system (CNS) diseases. Hypoxic treatment also increases the effectiveness of stem cell therapy. In this review, we firstly describe the role of hypoxia in different stem cells, such as embryonic stem cells, NSCs, and induced pluripotent stem cells, and discuss the role of hypoxia-treated stem cells in CNS diseases treatment. Furthermore, we highlight the role and mechanisms of hypoxia in regulating adult neurogenesis in the SVZ and DG and adult proliferation of other cells in the CNS.
Collapse
Affiliation(s)
- Gaifen Li
- Laboratory of Brain DisordersMinistry of Science and TechnologyCollaborative Innovation Center for Brain DisordersBeijing Institute of Brain DisordersCapital Medical UniversityBeijingChina
- Department of NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
| | - Jia Liu
- Laboratory of Brain DisordersMinistry of Science and TechnologyCollaborative Innovation Center for Brain DisordersBeijing Institute of Brain DisordersCapital Medical UniversityBeijingChina
| | - Yuying Guan
- Laboratory of Brain DisordersMinistry of Science and TechnologyCollaborative Innovation Center for Brain DisordersBeijing Institute of Brain DisordersCapital Medical UniversityBeijingChina
- Department of NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
| | - Xunming Ji
- Laboratory of Brain DisordersMinistry of Science and TechnologyCollaborative Innovation Center for Brain DisordersBeijing Institute of Brain DisordersCapital Medical UniversityBeijingChina
- Department of NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
27
|
Neuroligin-3 Regulates Excitatory Synaptic Transmission and EPSP-Spike Coupling in the Dentate Gyrus In Vivo. Mol Neurobiol 2021; 59:1098-1111. [PMID: 34845591 PMCID: PMC8857112 DOI: 10.1007/s12035-021-02663-9] [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: 06/08/2021] [Accepted: 11/22/2021] [Indexed: 11/27/2022]
Abstract
Neuroligin-3 (Nlgn3), a neuronal adhesion protein implicated in autism spectrum disorder (ASD), is expressed at excitatory and inhibitory postsynapses and hence may regulate neuronal excitation/inhibition balance. To test this hypothesis, we recorded field excitatory postsynaptic potentials (fEPSPs) in the dentate gyrus of Nlgn3 knockout (KO) and wild-type mice. Synaptic transmission evoked by perforant path stimulation was reduced in KO mice, but coupling of the fEPSP to the population spike was increased, suggesting a compensatory change in granule cell excitability. These findings closely resemble those in neuroligin-1 (Nlgn1) KO mice and could be partially explained by the reduction in Nlgn1 levels we observed in hippocampal synaptosomes from Nlgn3 KO mice. However, unlike Nlgn1, Nlgn3 is not necessary for long-term potentiation. We conclude that while Nlgn1 and Nlgn3 have distinct functions, both are required for intact synaptic transmission in the mouse dentate gyrus. Our results indicate that interactions between neuroligins may play an important role in regulating synaptic transmission and that ASD-related neuroligin mutations may also affect the synaptic availability of other neuroligins.
Collapse
|
28
|
Blockade of adenosine A 2A receptor alleviates cognitive dysfunction after chronic exposure to intermittent hypoxia in mice. Exp Neurol 2021; 350:113929. [PMID: 34813840 DOI: 10.1016/j.expneurol.2021.113929] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 10/25/2021] [Accepted: 11/16/2021] [Indexed: 11/22/2022]
Abstract
Obstructive sleep apnea-hypopnea syndrome (OSAHS) is widely known for its multiple systems damage, especially neurocognitive deficits in children. Since their discovery, adenosine A2A receptors (A2ARs) have been considered as key elements in signaling pathways mediating neurodegenerative diseases such as Huntington's and Alzheimer's, as well as cognitive function regulation. Herein, we investigated A2AR role in cognitive impairment induced by chronic intermittent hypoxia (CIH). Mice were exposed to CIH 7 h every day for 4 weeks, and intraperitoneally injected with A2AR agonist CGS21680 or A2AR antagonist SCH58261 half an hour before IH exposure daily. The 8-arm radial arm maze was utilized to assess spatial memory after CIH exposures.To validate findings using pharmacology, the impact of intermittent hypoxia was investigated in A2AR knockout mice. CIH-induced memory dysfunction was manifested by increased error rates in the radial arm maze test. The behavioral changes were associated with hippocampal pathology, neuronal apoptosis, and synaptic plasticity impairment. The stimulation of adenosine A2AR exacerbated memory impairment with more serious neuropathological damage, attenuated long-term potentiation (LTP), syntaxin down-regulation, and increased BDNF protein. Moreover, apoptosis-promoting protein cleaved caspase-3 was upregulated while anti-apoptotic protein Bcl-2 was downregulated. Consistent with these findings, A2AR inhibition with SCH58261 and A2AR deletion exhibited the opposite result. Overall, these findings suggest that A2AR plays a critical role in CIH-induced impairment of learning and memory by accelerating hippocampal neuronal apoptosis and reducing synaptic plasticity. Blockade of adenosine A2A receptor alleviates cognitive dysfunction after chronic exposure to intermittent hypoxia in mice.
Collapse
|
29
|
Framnes-DeBoer SN, Jones AA, Kang MY, Propsom K, Nelson LR, Arble DM. The timing of intermittent hypoxia differentially affects macronutrient intake and energy substrate utilization in mice. Am J Physiol Endocrinol Metab 2021; 321:E543-E550. [PMID: 34459217 DOI: 10.1152/ajpendo.00183.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sleep apnea is a common sleep disorder characterized by periodic breathing cessation and intermittent hypoxia (IH). Although previous studies have demonstrated that IH alone can influence metabolic outcomes such as body weight, it remains unclear how the timing of IH can specifically affect these outcomes. Here, we examine how pairing 10-h periods of IH to either the animals' resting phase (e.g., IH during the day) or active phase (e.g., IH during the night) differentially affects body weight, macronutrient selection, energy expenditure, respiratory exchange rate, and glucose tolerance. We find that in contrast to mice exposed to IH during the night, mice exposed to IH during the day preferentially decrease their carbohydrate intake and switch to fat metabolism. Moreover, when the IH stimulus was removed, mice that had been exposed to day IH continued to eat a minimal amount of carbohydrates and consumed a higher percentage of kilocalorie from fat for at least 5 days. These data demonstrate that food choice and substrate utilization are secondary to the timing of IH but not IH itself. Taken together, these data have key clinical implications for individuals with sleep apnea and particularly those who are also experiencing circadian disruption such as night-shift workers.NEW & NOTEWORTHY Pairing repeated hypoxic episodes to a mouse's resting phase during the day preferentially decreases carbohydrate intake and results in a switch to metabolic fat oxidation. These data indicate that the timing of intermittent hypoxia should be considered when calculating sleep apnea's effects on metabolic outcomes.
Collapse
Affiliation(s)
| | - Aaron A Jones
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin
| | - Michelle Y Kang
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin
| | - Kat Propsom
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin
| | - Lauren R Nelson
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin
| | - Deanna M Arble
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin
| |
Collapse
|
30
|
Chen L, Ren SY, Li RX, Liu K, Chen JF, Yang YJ, Deng YB, Wang HZ, Xiao L, Mei F, Wang F. Chronic Exposure to Hypoxia Inhibits Myelinogenesis and Causes Motor Coordination Deficits in Adult Mice. Neurosci Bull 2021; 37:1397-1411. [PMID: 34292513 PMCID: PMC8490606 DOI: 10.1007/s12264-021-00745-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 03/06/2021] [Indexed: 12/18/2022] Open
Abstract
Exposure to chronic hypoxia is considered to be a risk factor for deficits in brain function in adults, but the underlying mechanisms remain largely unknown. Since active myelinogenesis persists in the adult central nervous system, here we aimed to investigate the impact of chronic hypoxia on myelination and the related functional consequences in adult mice. Using a transgenic approach to label newly-generated myelin sheaths (NG2-CreERTM; Tau-mGFP), we found that myelinogenesis was highly active in most brain regions, such as the motor cortex and corpus callosum. After exposure to hypoxia (10% oxygen) 12 h per day for 4 weeks, myelinogenesis was largely inhibited in the 4-month old brain and the mice displayed motor coordination deficits revealed by the beam-walking test. To determine the relationship between the inhibited myelination and functional impairment, we induced oligodendroglia-specific deletion of the transcription factor Olig2 by tamoxifen (NG2-CreERTM; Tau-mGFP; Olig2 fl/fl) in adult mice to mimic the decreased myelinogenesis caused by hypoxia. The deletion of Olig2 inhibited myelinogenesis and consequently impaired motor coordination, suggesting that myelinogenesis is required for motor function in adult mice. To understand whether enhancing myelination could protect brain functions against hypoxia, we treated hypoxic mice with the myelination-enhancing drug-clemastine, which resulted in enhanced myelogenesis and improved motor coordination. Taken together, our data indicate that chronic hypoxia inhibits myelinogenesis and causes functional deficits in the brain and that enhancing myelinogenesis protects brain functions against hypoxia-related deficits.
Collapse
Affiliation(s)
- Lin Chen
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China
| | - Shu-Yu Ren
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China
| | - Rui-Xue Li
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China
| | - Kun Liu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China
| | - Jing-Fei Chen
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China
| | - Yu-Jian Yang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China
| | - Yong-Bin Deng
- Department of Neurosurgery, Chongqing Emergency Medical Center, Chongqing University, Chongqing, 400014, China
| | - Han-Zhi Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China
| | - Lan Xiao
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China
| | - Feng Mei
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China.
| | - Fei Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
31
|
Nanduri J, Wang N, Wang BL, Prabhakar NR. Lysine demethylase KDM6B regulates HIF-1α-mediated systemic and cellular responses to intermittent hypoxia. Physiol Genomics 2021; 53:385-394. [PMID: 34297635 PMCID: PMC8887999 DOI: 10.1152/physiolgenomics.00045.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Intermittent hypoxia (IH) is a hallmark manifestation of obstructive sleep apnea (OSA). Rodents treated with IH exhibit hypertension. Hypoxia-inducible factor (HIF)-1-dependent transcriptional activation of NADPH oxidases (Nox) and the resulting increase in reactive oxygen species (ROS) levels is a major molecular mechanism underlying IH/OSA-induced hypertension. Jumanji C (JmjC)-containing histone lysine demethylases (JmjC-KDMs) are coactivators of HIF-1-dependent transcriptional activation. In the present study, we tested the hypothesis that JmjC-KDMs are required for IH-evoked HIF-1 transcriptional activation of Nox4 and the ensuing hypertension. Studies were performed on pheochromocytoma (PC)12 cells and rats. IH increased KDM6B protein and enzyme activity in PC12 cells in an HIF-1-independent manner as evidenced by unaltered KDM6B activation by IH in HIF-1α shRNA-treated cells. Cells treated with IH showed increased HIF-1-dependent Nox4 transcription as indicated by increased HIF-1α binding to hypoxia-responsive element (HRE) sequence of the Nox4 gene promoter demonstrated by chromatin immunoprecipitation (ChiP) assay. Pharmacological blockade of KDM6B with GSKJ4, a specific KDM6 inhibitor, or genetic silencing of KDM6B with shRNA abolished IH-induced Nox4 transcriptional activation by blocking HIF-1α binding to the promoter of the Nox4 gene. Treating IH-exposed rats with GSKJ4 showed: 1) absence of KDM6B activation and HIF-1-dependent Nox4 transcription in the adrenal medullae, and 2) absence of elevated plasma catecholamines and hypertension. Collectively, these findings indicate that KDM6B functions as a coactivator of HIF-1-mediated Nox4 transactivation and demonstrates a hitherto uncharacterized role for KDMs in IH-induced hypertension by HIF-1.
Collapse
Affiliation(s)
- Jayasri Nanduri
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, The University of Chicago, Chicago, Illinois
| | - Ning Wang
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, The University of Chicago, Chicago, Illinois
| | - Benjamin L. Wang
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, The University of Chicago, Chicago, Illinois
| | - Nanduri R. Prabhakar
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, The University of Chicago, Chicago, Illinois
| |
Collapse
|
32
|
Involvement of Hepcidin in Cognitive Damage Induced by Chronic Intermittent Hypoxia in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8520967. [PMID: 34394834 PMCID: PMC8357469 DOI: 10.1155/2021/8520967] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/03/2021] [Accepted: 07/17/2021] [Indexed: 02/07/2023]
Abstract
Obstructive sleep apnea (OSA) patients exhibit different degrees of cognitive impairment, which is related to the activation of reactive oxygen species (ROS) production by chronic intermittent hypoxia (CIH) and the deposition of iron in the brain. As a central regulator of iron homeostasis, whether hepcidin is involved in OSA-induced cognitive impairment has not been clarified. In order to simulate OSA, we established the mouse model by reducing the percentage of inspired O2 (FiO2) from 21% to 5%, 20 times/h for 8 h/day. We found hepcidin was rising during CIH, along with increasing iron levels and neuron loss. Then, we constructed a mouse with astrocyte-specific knockdown hepcidin gene (shHamp). During CIH exposure, the shHamp mice showed a lower level of total iron and neuronal iron in the hippocampus, via stabilizing ferroportin 1 (FPN1) and decreasing L-ferritin (FTL) levels, when compared with wild-type (WT) mice. Furthermore, the shHamp mice showed a decrease of ROS by downregulating the elevated NADPH oxidase (NOX2) and 4-hydroxynonenal (4-HNE) levels mediated by CIH. In addition, the shHamp mice presented improved cognitive deficit by improving synaptic plasticity and BDNF expression in the hippocampus when subjected to CIH. Therefore, our data revealed that highly expressed hepcidin might promote the degradation of FPN1, resulting in neuronal iron deposition, oxidative stress damage, reduced synaptic plasticity, and impaired cognitive performance during CIH exposure.
Collapse
|
33
|
Intermittent Hypoxia and Effects on Early Learning/Memory: Exploring the Hippocampal Cellular Effects of Pediatric Obstructive Sleep Apnea. Anesth Analg 2021; 133:93-103. [PMID: 33234943 DOI: 10.1213/ane.0000000000005273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review provides an update on the neurocognitive phenotype of pediatric obstructive sleep apnea (OSA). Pediatric OSA is associated with neurocognitive deficits involving memory, learning, and executive functioning. Adenotonsillectomy (AT) is presently accepted as the first-line surgical treatment for pediatric OSA, but the executive function deficits do not resolve postsurgery, and the timeline for recovery remains unknown. This finding suggests that pediatric OSA potentially causes irreversible damage to multiple areas of the brain. The focus of this review is the hippocampus, 1 of the 2 major sites of postnatal neurogenesis, where new neurons are formed and integrated into existing circuitry and the mammalian center of learning/memory functions. Here, we review the clinical phenotype of pediatric OSA, and then discuss existing studies of OSA on different cell types in the hippocampus during critical periods of development. This will set the stage for future study using preclinical models to understand the pathogenesis of persistent neurocognitive dysfunction in pediatric OSA.
Collapse
|
34
|
Arias-Cavieres A, Fonteh A, Castro-Rivera CI, Garcia AJ. Intermittent Hypoxia causes targeted disruption to NMDA receptor dependent synaptic plasticity in area CA1 of the hippocampus. Exp Neurol 2021; 344:113808. [PMID: 34256046 DOI: 10.1016/j.expneurol.2021.113808] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/21/2021] [Accepted: 07/08/2021] [Indexed: 12/27/2022]
Abstract
Changed NMDA receptor (NMDAr) physiology is implicated with cognitive deficit resulting from conditions ranging from normal aging to neurological disease. Using intermittent hypoxia (IH) to experimentally model untreated sleep apnea, a clinical condition whose comorbidities include neurocognitive impairment, we recently demonstrated that IH causes a pro-oxidant condition that contributes to deficits in spatial memory and in NMDAr-dependent long-term potentiation (LTP). However, the impact of IH on additional forms of synaptic plasticity remains ill-defined. Here we show that IH prevents the induction of NMDAr-dependent LTP and long-term depression (LTD) in hippocampal brain slices from mice exposed to ten days of IH (IH10) yet spares NMDAr-independent forms of synaptic plasticity. Deficits in synaptic plasticity were accompanied by a reduction in hippocampal GluN1 expression. Acute manipulation of redox state using the reducing agent, Dithiothreitol (DTT) stimulated the NMDAr-dependent fEPSP following IH10. However, acute use of either DTT or MnTMPyP did not restore NMDAr-dependent synaptic plasticity after IH10 or prevent the IH-dependent reduction in GluN1, the obligatory subunit of the NMDAr. In contrast, MnTMPyP during IH10 (10-MnTMPyP), prevented the suppressive effects of IH on both NMDAr-dependent synaptic plasticity and GluN1 expression. These findings indicate that while the IH-dependent pro-oxidant state causes reversible oxidative neuromodulation of NMDAr activity, acute manipulation of redox state is ineffective in rescuing two key effects of IH related to the NMDAr within the hippocampus. These IH-dependent changes associated with the NMDAr may be a primary avenue by which IH enhances the vulnerability to impaired learning and memory when sleep apnea is left untreated in normal aging and in disease.
Collapse
Affiliation(s)
- Alejandra Arias-Cavieres
- Institute for Integrative Physiology, The University of Chicago, USA; Department of Medicine, Section of Emergency Medicine, The University of Chicago, USA
| | - Ateh Fonteh
- Department of Medicine, Section of Emergency Medicine, The University of Chicago, USA
| | - Carolina I Castro-Rivera
- Institute for Integrative Physiology, The University of Chicago, USA; Grossman Institute for Neuroscience, Quantitative Biology & Human Behavior, The University of Chicago, USA
| | - Alfredo J Garcia
- Institute for Integrative Physiology, The University of Chicago, USA; Grossman Institute for Neuroscience, Quantitative Biology & Human Behavior, The University of Chicago, USA; Department of Medicine, Section of Emergency Medicine, The University of Chicago, USA.
| |
Collapse
|
35
|
Sefiani A, Geoffroy CG. The Potential Role of Inflammation in Modulating Endogenous Hippocampal Neurogenesis After Spinal Cord Injury. Front Neurosci 2021; 15:682259. [PMID: 34220440 PMCID: PMC8249862 DOI: 10.3389/fnins.2021.682259] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022] Open
Abstract
Currently there are approximately 291,000 people suffering from a spinal cord injury (SCI) in the United States. SCI is associated with traumatic changes in mobility and neuralgia, as well as many other long-term chronic health complications, including metabolic disorders, diabetes mellitus, non-alcoholic steatohepatitis, osteoporosis, and elevated inflammatory markers. Due to medical advances, patients with SCI survive much longer than previously. This increase in life expectancy exposes them to novel neurological complications such as memory loss, cognitive decline, depression, and Alzheimer's disease. In fact, these usually age-associated disorders are more prevalent in people living with SCI. A common factor of these disorders is the reduction in hippocampal neurogenesis. Inflammation, which is elevated after SCI, plays a major role in modulating hippocampal neurogenesis. While there is no clear consensus on the mechanism of the decline in hippocampal neurogenesis and cognition after SCI, we will examine in this review how SCI-induced inflammation could modulate hippocampal neurogenesis and provoke age-associated neurological disorders. Thereafter, we will discuss possible therapeutic options which may mitigate the influence of SCI associated complications on hippocampal neurogenesis.
Collapse
|
36
|
Luijerink LLM, Vivekanandarajah A, Waters KA, Machaalani R. The α7 and β2 nicotinic acetylcholine receptor subunits regulate apoptosis in the infant hippocampus, and in sudden infant death syndrome (SIDS). Apoptosis 2021; 25:574-589. [PMID: 32577853 DOI: 10.1007/s10495-020-01618-0] [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] [Indexed: 12/16/2022]
Abstract
Apoptosis is increased in the hippocampus of infants who died of sudden infant death syndrome (SIDS), yet it is not known via which mechanism this has occurred. Following existing support for a role of the α7 and β2 nicotinic acetylcholine receptor (nAChR) subunits in apoptotic regulation, we aimed to determine whether these subunits are altered in the SIDS hippocampus and if they are correlated with cell death markers of active caspase-3 (Casp-3) and TUNEL. Further analyses were run according to the presence of major SIDS risk factors related to hypoxia (bed-sharing and prone sleeping), infection (presence of an upper respiratory tract infection (URTI)), cigarette smoke exposure and gender. Immunohistochemical expression of the markers was studied in 4 regions of the hippocampus (Cornu Ammonis (CA)1, CA2, CA3, CA4) and subiculum amongst 52 infants (aged 1-7 months) who died suddenly and unexpectedly (SUDI) and for whom the cause of death was explained (eSUDI; n = 9), or not and characterised as SIDS I (n = 8) and SIDS II (n = 35) according to the San Diego diagnostic criteria. Results showed that SIDS II infants had widespread increases in TUNEL compared with eSUDI and SIDS I infants, as well as increased α7 and Casp-3 in CA2 compared to eSUDI infants, although these changes were predominant amongst infants who did not bed-share. Cigarette smoke exposure had minimal effects on the markers, while an URTI was associated with changes in all markers (after accounting for bed-sharing). Our findings support the role of nAChRs in regulating apoptosis in the SIDS hippocampus, and highlight the need for separate analysis according to risk factors.
Collapse
Affiliation(s)
- L L M Luijerink
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Medical Foundation Building K25, Sydney, NSW, 2006, Australia
| | - A Vivekanandarajah
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Medical Foundation Building K25, Sydney, NSW, 2006, Australia
| | - K A Waters
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Medical Foundation Building K25, Sydney, NSW, 2006, Australia.,The Children's Hospital at Westmead, Westmead, NSW, 2146, Australia
| | - R Machaalani
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Medical Foundation Building K25, Sydney, NSW, 2006, Australia. .,The Children's Hospital at Westmead, Westmead, NSW, 2146, Australia.
| |
Collapse
|
37
|
Butt UJ, Hassouna I, Fernandez Garcia-Agudo L, Steixner-Kumar AA, Depp C, Barnkothe N, Zillmann MR, Ronnenberg A, Bonet V, Goebbels S, Nave KA, Ehrenreich H. CaMKIIα Expressing Neurons to Report Activity-Related Endogenous Hypoxia upon Motor-Cognitive Challenge. Int J Mol Sci 2021; 22:3164. [PMID: 33804598 PMCID: PMC8003772 DOI: 10.3390/ijms22063164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/16/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
Abstract
We previously introduced the brain erythropoietin (EPO) circle as a model to explain the adaptive 'brain hardware upgrade' and enhanced performance. In this fundamental circle, brain cells, challenged by motor-cognitive tasks, experience functional hypoxia, triggering the expression of EPO among other genes. We attested hypoxic cells by a transgenic reporter approach under the ubiquitous CAG promoter, with Hif-1α oxygen-dependent degradation-domain (ODD) fused to CreERT2-recombinase. To specifically focus on the functional hypoxia of excitatory pyramidal neurons, here, we generated CaMKIIα-CreERT2-ODD::R26R-tdTomato mice. Behavioral challenges, light-sheet microscopy, immunohistochemistry, single-cell mRNA-seq, and neuronal cultures under normoxia or hypoxia served to portray these mice. Upon complex running wheel performance as the motor-cognitive task, a distinct increase in functional hypoxic neurons was assessed immunohistochemically and confirmed three-dimensionally. In contrast, fear conditioning as hippocampal stimulus was likely too short-lived to provoke neuronal hypoxia. Transcriptome data of hippocampus under normoxia versus inspiratory hypoxia revealed increases in CA1 CaMKIIα-neurons with an immature signature, characterized by the expression of Dcx, Tbr1, CaMKIIα, Tle4, and Zbtb20, and consistent with accelerated differentiation. The hypoxia reporter response was reproduced in vitro upon neuronal maturation. To conclude, task-associated activity triggers neuronal functional hypoxia as a local and brain-wide reaction mediating adaptive neuroplasticity. Hypoxia-induced genes such as EPO drive neuronal differentiation, brain maturation, and improved performance.
Collapse
Affiliation(s)
- Umer Javed Butt
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; (U.J.B.); (I.H.); (L.F.G.-A.); (A.A.S.-K.); (N.B.); (M.R.Z.); (A.R.); (V.B.)
| | - Imam Hassouna
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; (U.J.B.); (I.H.); (L.F.G.-A.); (A.A.S.-K.); (N.B.); (M.R.Z.); (A.R.); (V.B.)
| | - Laura Fernandez Garcia-Agudo
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; (U.J.B.); (I.H.); (L.F.G.-A.); (A.A.S.-K.); (N.B.); (M.R.Z.); (A.R.); (V.B.)
| | - Agnes A. Steixner-Kumar
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; (U.J.B.); (I.H.); (L.F.G.-A.); (A.A.S.-K.); (N.B.); (M.R.Z.); (A.R.); (V.B.)
| | - Constanze Depp
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; (C.D.); (S.G.); (K.-A.N.)
| | - Nadine Barnkothe
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; (U.J.B.); (I.H.); (L.F.G.-A.); (A.A.S.-K.); (N.B.); (M.R.Z.); (A.R.); (V.B.)
| | - Matthias R. Zillmann
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; (U.J.B.); (I.H.); (L.F.G.-A.); (A.A.S.-K.); (N.B.); (M.R.Z.); (A.R.); (V.B.)
| | - Anja Ronnenberg
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; (U.J.B.); (I.H.); (L.F.G.-A.); (A.A.S.-K.); (N.B.); (M.R.Z.); (A.R.); (V.B.)
| | - Viktoria Bonet
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; (U.J.B.); (I.H.); (L.F.G.-A.); (A.A.S.-K.); (N.B.); (M.R.Z.); (A.R.); (V.B.)
| | - Sandra Goebbels
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; (C.D.); (S.G.); (K.-A.N.)
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; (C.D.); (S.G.); (K.-A.N.)
| | - Hannelore Ehrenreich
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; (U.J.B.); (I.H.); (L.F.G.-A.); (A.A.S.-K.); (N.B.); (M.R.Z.); (A.R.); (V.B.)
| |
Collapse
|
38
|
Khuu MA, Nallamothu T, Castro-Rivera CI, Arias-Cavieres A, Szujewski CC, Garcia Iii AJ. Stage-dependent effects of intermittent hypoxia influence the outcome of hippocampal adult neurogenesis. Sci Rep 2021; 11:6005. [PMID: 33727588 PMCID: PMC7966401 DOI: 10.1038/s41598-021-85357-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 02/25/2021] [Indexed: 12/14/2022] Open
Abstract
Over one billion adults worldwide are estimated to suffer from sleep apnea, a condition with wide-reaching effects on brain health. Sleep apnea causes cognitive decline and is a risk factor for neurodegenerative conditions such as Alzheimer’s disease. Rodents exposed to intermittent hypoxia (IH), a hallmark of sleep apnea, exhibit spatial memory deficits associated with impaired hippocampal neurophysiology and dysregulated adult neurogenesis. We demonstrate that IH creates a pro-oxidant condition that reduces the Tbr2+ neural progenitor pool early in the process, while also suppressing terminal differentiation of adult born neurons during late adult neurogenesis. We further show that IH-dependent cell-autonomous hypoxia inducible factor 1-alpha (HIF1a) signaling is activated in early neuroprogenitors and enhances the generation of adult born neurons upon termination of IH. Our findings indicate that oscillations in oxygen homeostasis, such as those found in sleep apnea, have complex stage-dependent influence over hippocampal adult neurogenesis.
Collapse
Affiliation(s)
- Maggie A Khuu
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA
| | - Thara Nallamothu
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA
| | - Carolina I Castro-Rivera
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA.,Committee On Neurobiology, The University of Chicago, Chicago, IL, 60307, USA.,Grossman Institute for Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, 60637, USA
| | - Alejandra Arias-Cavieres
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA
| | - Caroline C Szujewski
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA.,Committee On Neurobiology, The University of Chicago, Chicago, IL, 60307, USA.,Grossman Institute for Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, 60637, USA
| | - Alfredo J Garcia Iii
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA. .,Committee On Neurobiology, The University of Chicago, Chicago, IL, 60307, USA. .,Grossman Institute for Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
39
|
Hernández-Soto R, Villasana-Salazar B, Pinedo-Vargas L, Peña-Ortega F. Chronic intermittent hypoxia alters main olfactory bulb activity and olfaction. Exp Neurol 2021; 340:113653. [PMID: 33607078 DOI: 10.1016/j.expneurol.2021.113653] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 02/02/2021] [Accepted: 02/14/2021] [Indexed: 02/08/2023]
Abstract
Olfactory dysfunction is commonly observed in patients with obstructive sleep apnea (OSA), which is related to chronic intermittent hypoxia (CIH). OSA patients exhibit alterations in discrimination, identification and odor detection threshold. These olfactory functions strongly rely on neuronal processing within the main olfactory bulb (MOB). However, a direct evaluation of the effects of controlled CIH on olfaction and MOB network activity has not been performed. Here, we used electrophysiological field recordings in vivo to evaluate the effects of 21-day-long CIH on MOB network activity and its response to odors. In addition, we assessed animals´ olfaction with the buried food and habituation/dishabituation tests. We found that mice exposed to CIH show alterations in MOB spontaneous activity in vivo, consisting of a reduction in beta and gamma frequency bands power along with an increase in the theta band power. Likewise, the MOB was less responsive to odor stimulation, since the proportional increase of the power of its population activity in response to four different odorants was smaller than the one observed in control animals. These CIH-induced MOB functional alterations correlate with a reduction in the ability to detect, habituate and discriminate olfactory stimuli. Our findings indicate that CIH generates alterations in the MOB neural network, which could be involved in the olfactory deterioration in patients with OSA.
Collapse
Affiliation(s)
- Rebeca Hernández-Soto
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Querétaro, Mexico
| | - Benjamín Villasana-Salazar
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Querétaro, Mexico
| | - Laura Pinedo-Vargas
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Querétaro, Mexico
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Querétaro, Mexico.
| |
Collapse
|
40
|
Shen H, Meng Y, Liu D, Qin Z, Huang H, Pan L, Wang W, Kang J. α7 Nicotinic Acetylcholine Receptor Agonist PNU-282987 Ameliorates Cognitive Impairment Induced by Chronic Intermittent Hypoxia. Nat Sci Sleep 2021; 13:579-590. [PMID: 34007230 PMCID: PMC8123952 DOI: 10.2147/nss.s296701] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 04/13/2021] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Cognitive impairment is an important complication of obstructive sleep apnea (OSA). Chronic intermittent hypoxia (CIH), the main pathophysiological characteristics of OSA, is closely related to cognitive dysfunction and may be mediated by alpha-7 nicotinic acetylcholine receptors (α7nAChR). This study investigated the effects and clarified the mechanisms of α7nAChR on the cognitive function of mice with CIH. METHODS Thirty CD-1 mice were randomly divided into room air (RA), CIH-2 weeks (CIH2W), and CIH-4 weeks (CIH4W) groups. Cognitive function was evaluated by novel object recognition (NOR) and Morris water maze (MWM) tests after exposure. Then, 104 CD-1 mice were exposed to CIH for 4 weeks and randomly divided into four groups: CIH4W (control), with dimethyl sulfoxide (DMSO) (sham), with α7nAChR-specific agonist PNU-282987 (PNU), and with α7nAChR-specific inhibitor methyllycaconitine and PNU-282987 (MLA+PNU). In addition to the evaluation of cognitive function, apoptotic bodies in the hippocampus were detected by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, changes in p-CREB and BDNF were detected by immunohistochemistry, while those of ERK1/2, CREB, PGC-1α, FNDC5, and BDNF were detected by Western blotting in the hippocampal tissues of the mice. RESULTS Compared to the CIH2W and RA groups, the CIH4W group showed cognitive dysfunction in the NOR and MWM tests. The changes in cognitive dysfunction were alleviated by PNU-282987; furthermore, MLA pretreatment offset the effect. In hippocampal tissues, TUNEL assays showed decreased apoptotic cells, immunohistochemical staining showed increased expressions of p-CREB and BDNF. The expression levels of p-ERK1/2/t-ERK1/2, p-CREB/t-CREB, PGC-1α, FNDC5, and BDNF were increased after PNU-282987 injection. CONCLUSION Four weeks of CIH caused cognitive dysfunction in mice. Activating α7nAChR might ameliorate this dysfunction by upregulating the ERK1/2/CREB signaling pathway; enhancing PGC-1α, FNDC5, and BDNF expression levels; and reducing cell apoptosis in the hippocampal tissue of mice.
Collapse
Affiliation(s)
- Hui Shen
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yanling Meng
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Dan Liu
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Zheng Qin
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Hong Huang
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Lei Pan
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Wei Wang
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Jian Kang
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
41
|
Chao B, Zhang L, Pan J, Zhang Y, Chen Y, Xu M, Huang S. Stanniocalcin-1 Overexpression Prevents Depression-Like Behaviors Through Inhibition of the ROS/NF-κB Signaling Pathway. Front Psychiatry 2021; 12:644383. [PMID: 34194345 PMCID: PMC8238083 DOI: 10.3389/fpsyt.2021.644383] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/26/2021] [Indexed: 01/29/2023] Open
Abstract
Background: Depression is a burdensome psychiatric disorder presenting with disordered inflammation and neural plasticity. We conducted this study with an aim to explore the effect of stanniocalcin-1 (STC1) on inflammation and neuron injury in rats with depression-like behaviors. Methods: A model of depression-like behaviors was established in Wistar rats by stress stimulation. Adeno-associated virus (AAV)-packaged STC1 overexpression sequence or siRNA against STC1 was introduced into rats to enhance or silence the STC1 expression. Moreover, we measured pro-inflammatory and anti-inflammatory proteins, superoxide dismutase (SOD), catalase (CAT), malondialdehyde (MDA) and reactive oxygen species (ROS) production. An in vitro model was induced in hippocampal neurons by CORT to explore the effect of STC1 on the neuron viability, toxicity and apoptosis. RT-qPCR and Western blot assay were employed to determine the expression of STC1 and nuclear factor κB (NF-κB) signaling pathway-related genes. Results: STC1 was under-expressed in the hippocampus of rats with depression-like behaviors, while its overexpression could reduce the depression-like behaviors in the stress-stimulated rats. Furthermore, overexpression of STC1 resulted in enhanced neural plasticity, reduced release of pro-inflammatory proteins, elevated SOD and CAT and diminished MDA level in the hippocampus of rats with depression-like behaviors. Overexpressed STC1 blocked the ROS/NF-κB signaling pathway, thereby enhancing the viability of CORT-treated neurons while repressing their toxicity and apoptosis. Conclusion: Collectively, overexpression of STC1 inhibits inflammation and protects neuron injury in rats with depression-like behaviors by inactivating the ROS/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Bin Chao
- Traditional Chinese Medicine Research and Development Center, Guang'anmen Hospital, China Academy of Chinese Medicine, Beijing, China
| | - Lili Zhang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medicine, Beijing, China
| | - Juhua Pan
- Traditional Chinese Medicine Research and Development Center, Guang'anmen Hospital, China Academy of Chinese Medicine, Beijing, China
| | - Ying Zhang
- Traditional Chinese Medicine Research and Development Center, Guang'anmen Hospital, China Academy of Chinese Medicine, Beijing, China
| | - Yuxia Chen
- Traditional Chinese Medicine Research and Development Center, Guang'anmen Hospital, China Academy of Chinese Medicine, Beijing, China
| | - Manman Xu
- Traditional Chinese Medicine Research and Development Center, Guang'anmen Hospital, China Academy of Chinese Medicine, Beijing, China
| | - Shijing Huang
- Traditional Chinese Medicine Research and Development Center, Guang'anmen Hospital, China Academy of Chinese Medicine, Beijing, China
| |
Collapse
|
42
|
Zhou L, Liu G, Luo H, Li H, Peng Y, Zong D, Ouyang R. Aberrant Hippocampal Network Connectivity Is Associated With Neurocognitive Dysfunction in Patients With Moderate and Severe Obstructive Sleep Apnea. Front Neurol 2020; 11:580408. [PMID: 33362692 PMCID: PMC7759642 DOI: 10.3389/fneur.2020.580408] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/11/2020] [Indexed: 01/20/2023] Open
Abstract
Objectives: This work aims to explore the changes of functional connectivity (FC) within the hippocampus network in patients with moderate and severe obstructive sleep apnea (OSA) and its correlation with neurocognitive dysfunction to explore the potential neurophysiological mechanism. Methods: A total of 32 treatment-naïve patients with moderate or severe OSA and 26 healthy controls (HCs), matched in age, gender, and education, underwent the evaluations of Epworth Sleep Scale, neurocognitive function, full-night polysomnography, and resting-state functional magnetic resonance imaging. The FC map of the hippocampus to other brain areas was compared among 15 OSA patients and 15 HCs with little head motion. Finally, the correlation between hippocampus FC strength and respiratory sleep parameters and neurocognitive assessments was analyzed. Results: Compared with HCs, the right hippocampus showed a significantly decreased FC with the bilateral insular lobe, right thalamus, and right anterior cingulate gyrus (ACG) and an increased FC with the right superior and middle temporal gyrus, left posterior cingulate gyrus, and left angular gyrus in the patients with OSA. The left hippocampus presented a significantly decreased FC with the left anterior cerebellum in patients with OSA. In addition, the aberrant right hippocampal FC with the right ACG was significantly correlated with disease severity and disrupted sleep architecture in the OSA group. Furthermore, after adjusting the related confounding factors, the FC strength between the right hippocampus, right insular lobe, and right thalamus was positively associated with the scores of Stroop Color-Word Test (SCWT) or Hopkins Verbal Learning Test-Revised (HVLT-R), while the FC between the right hippocampus and the right middle temporal gyrus was negatively correlated with the scores of HVLT-R. The right hippocampus FC with right superior temporal gyrus, left angular gyrus, and ACG were all negatively related to the scores of the symbol coding test (r = -0.642, p = 0.045; r = -0.638, p = 0.047; r = -0.753, p = 0.012), respectively. The FC between the left hippocampal and the left anterior cerebellar lobe showed a positive relationship with the scores of HVLT-R (r = 0.757, p = 0.011) and CPT-3D (r = -0.801, p = 0.005). Conclusion: The hippocampus presented abnormal FC with the cerebral and cerebellar regions extensively in OSA, and the correlation between abnormal hippocampal network FC and neurocognitive dysfunction in OSA suggests a promising insight to explore the potential biomarker and pathophysiologic mechanism of neurocognitive dysfunction of OSA.
Collapse
Affiliation(s)
- Li Zhou
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Guiqian Liu
- Hunan Province Prevention and Treatment Institute for Occupational Diseases, Changsha, China
| | - Hong Luo
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Huabing Li
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yating Peng
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Dandan Zong
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Ruoyun Ouyang
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| |
Collapse
|
43
|
Neurocognitive and Synaptic Potentiation Deficits Are Mitigated by Inhibition of HIF1a Signaling following Intermittent Hypoxia in Rodents. eNeuro 2020; 7:7/6/ENEURO.0449-20.2020. [PMID: 33273035 PMCID: PMC7716431 DOI: 10.1523/eneuro.0449-20.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Highlighted Research Paper:A HIF1a-Dependent Pro-Oxidant State Disrupts Synaptic Plasticity and Impairs Spatial Memory in Response to Intermittent Hypoxia. Alejandra Arias-Cavieres, Maggie A. Khuu, Chinwendu U. Nwakudu, Jasmine E. Barnard, Gokhan Dalgin and Alfredo J. Garcia III
Collapse
|
44
|
Ramirez JM, Karlen-Amarante M, Wang JDJ, Bush NE, Carroll MS, Weese-Mayer DE, Huff A. The Pathophysiology of Rett Syndrome With a Focus on Breathing Dysfunctions. Physiology (Bethesda) 2020; 35:375-390. [PMID: 33052774 PMCID: PMC7864239 DOI: 10.1152/physiol.00008.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/21/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023] Open
Abstract
Rett syndrome (RTT), an X-chromosome-linked neurological disorder, is characterized by serious pathophysiology, including breathing and feeding dysfunctions, and alteration of cardiorespiratory coupling, a consequence of multiple interrelated disturbances in the genetic and homeostatic regulation of central and peripheral neuronal networks, redox state, and control of inflammation. Characteristic breath-holds, obstructive sleep apnea, and aerophagia result in intermittent hypoxia, which, combined with mitochondrial dysfunction, causes oxidative stress-an important driver of the clinical presentation of RTT.
Collapse
Affiliation(s)
- Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, Washington
- Departments of Neurological Surgery and Pediatrics, University of Washington School of Medicine, Seattle, Washington
| | - Marlusa Karlen-Amarante
- Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, Washington
- Department of Physiology and Pathology, School of Dentistry of Araraquara, São Paulo State University (UNESP), Araraquara, Brazil
| | - Jia-Der Ju Wang
- Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, Washington
| | - Nicholas E Bush
- Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, Washington
| | - Michael S Carroll
- Data Analytics and Reporting, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of Autonomic Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Debra E Weese-Mayer
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of Autonomic Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Alyssa Huff
- Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
45
|
Huang L, Wan Y, Dang Z, Yang P, Yang Q, Wu S. Hypoxic preconditioning ameliorated neuronal injury after middle cerebral artery occlusion by promoting neurogenesis. Brain Behav 2020; 10:e01804. [PMID: 32841552 PMCID: PMC7559635 DOI: 10.1002/brb3.1804] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/27/2020] [Accepted: 08/01/2020] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Sequelae of stroke were mainly caused by neuronal injury. Oxygen is a key factor affecting the microenvironment of neural stem cells (NSCs), and oxygen levels are used to promote NSC neurogenesis. In this study, effects of intermittent hypoxic preconditioning (HPC) on neurogenesis were investigated in a rat model of middle cerebral artery occlusion (MCAO). METHODS SD rats were used to establish the MCAO model. Nissl staining and Golgi staining were used to confirm the neuronal injury status in the MCAO model. Immunofluorescence, transmission electron microscopy, Western blot, and qPCR were used to observe the effects of HPC on neurogenesis. At the same time, the hypothesis that HPC could affect proliferation, apoptosis, differentiation, and migration of NSC was verified in vitro. RESULTS Hypoxic preconditioning significantly ameliorated the neuronal injury induced by MCAO. Compared with MCAO group, the dendrites, Edu+ /SOX2+ , Edu+ /DCX+ , Edu+ /NeuN+ , Edu+ /GFAP+ , and Edu+ /Tubulin+ positive cells in the HPC + MCAO group exhibited significantly difference. Similarly, axonal and other neuronal injuries in the HPC + MCAO group were also ameliorated. In the in vitro experiments, mild HPC significantly enhanced the viability of NSCs, promoted the migration of differentiated cells, and reduced apoptosis. CONCLUSIONS Our results showed that HPC significantly promotes neurogenesis after MCAO and ameliorates neuronal injury.
Collapse
Affiliation(s)
- Lu Huang
- Research Center for High Altitude Medicine, Qinghai University, Xining, China.,Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Xining, China.,Qinghai Provincial People's Hospital, Xining, China
| | - Yaqi Wan
- Qinghai Provincial People's Hospital, Xining, China
| | - Zhancui Dang
- Qinghai University Medical College, Xining, China
| | - Peng Yang
- Qinghai Provincial People's Hospital, Xining, China
| | - Quanyu Yang
- Qinghai University Medical College, Xining, China
| | - Shizheng Wu
- Qinghai Provincial People's Hospital, Xining, China
| |
Collapse
|
46
|
Villasana-Salazar B, Hernández-Soto R, Guerrero-Gómez ME, Ordaz B, Manrique-Maldonado G, Salgado-Puga K, Peña-Ortega F. Chronic intermittent hypoxia transiently increases hippocampal network activity in the gamma frequency band and 4-Aminopyridine-induced hyperexcitability in vitro. Epilepsy Res 2020; 166:106375. [PMID: 32745888 DOI: 10.1016/j.eplepsyres.2020.106375] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/21/2020] [Accepted: 05/28/2020] [Indexed: 12/12/2022]
Abstract
Chronic intermittent hypoxia (CIH) is the most distinct feature of obstructive sleep apnea (OSA), a common breathing and sleep disorder that leads to several neuropathological consequences, including alterations in the hippocampal network and in seizure susceptibility. However, it is currently unknown whether these alterations are permanent or remit upon normal oxygenation. Here, we investigated the effects of CIH on hippocampal spontaneous network activity and hyperexcitability in vitro and explored whether these alterations endure or fade after normal oxygenation. Results showed that applying CIH for 21 days to adult rats increases gamma-band hippocampal network activity and aggravates 4-Aminopyridine-induced epileptiform activity in vitro. Interestingly, these CIH-induced alterations remit after 30 days of normal oxygenation. Our findings indicate that hippocampal network alterations and increased seizure susceptibility induced by CIH are not permanent and can be spontaneously reverted, suggesting that therapeutic interventions against OSA in patients with epilepsy, such as surgery or continuous positive airway pressure (CPAP), could be favorable for seizure control.
Collapse
Affiliation(s)
- Benjamín Villasana-Salazar
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO 76230, México
| | - Rebeca Hernández-Soto
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO 76230, México
| | - María Estefanía Guerrero-Gómez
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO 76230, México
| | - Benito Ordaz
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO 76230, México
| | - Guadalupe Manrique-Maldonado
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO 76230, México
| | - Karla Salgado-Puga
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO 76230, México
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO 76230, México.
| |
Collapse
|
47
|
Zhang Y, Cao H, Qiu X, Xu D, Chen Y, Barnes GN, Tu Y, Gyabaah AT, Gharbal AHAA, Peng C, Cai J, Cai X. Neuroprotective Effects of Adenosine A1 Receptor Signaling on Cognitive Impairment Induced by Chronic Intermittent Hypoxia in Mice. Front Cell Neurosci 2020; 14:202. [PMID: 32733207 PMCID: PMC7363980 DOI: 10.3389/fncel.2020.00202] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/09/2020] [Indexed: 12/18/2022] Open
Abstract
Obstructive sleep apnea-hypopnea syndrome (OSAHS) is a breathing disorder associated with cognitive impairment. However, the mechanisms leading to cognitive deficits in OSAHS remain uncertain. In this study, a mouse model of chronic intermittent hypoxia (CIH) exposures were applied for simulating the deoxygenation-reoxygenation events occurring in OSAHS. The conventional adenosine A1 receptor gene (A1R) knockout mice and the A1R agonist CCPA- or antagonist DPCPX-administrated mice were utilized to determine the precise function of A1R signaling in the process of OSAHS-relevant cognitive impairment. We demonstrated that CIH induced morphological changes and apoptosis in hippocampal neurons. Further, CIH blunted hippocampal long-term potentiation (LTP) and resulted in learning/memory impairment. Disruption of adenosine A1R exacerbated morphological, cellular, and functional damage induced by CIH. In contrast, activation of adenosine A1R signaling reduced morphological changes and apoptosis of hippocampal neurons, promoted LTP, and enhanced learning and memory. A1Rs may up-regulate protein kinase C (PKC) and its subtype PKC-ζ through the activation of Gα(i) improve spatial learning and memory disorder induced by CIH in mice. Taken together, A1R signaling plays a neuroprotective role in CIH-induced cognitive dysfunction and pathological changes in the hippocampus.
Collapse
Affiliation(s)
- Yichun Zhang
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hongchao Cao
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China.,Department of Internal Medicine, Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No. 2 Hospital), Ningbo, China
| | - Xuehao Qiu
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Danfen Xu
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yifeng Chen
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Gregory N Barnes
- Department of Neurology, University of Louisville School of Medicine, Louisville, KY, United States.,Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, KY, United States
| | - Yunjia Tu
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Adwoa Takyiwaa Gyabaah
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | | | - Chenlei Peng
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China.,Department of Internal Medicine, Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No. 2 Hospital), Ningbo, China
| | - Jun Cai
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, KY, United States
| | - Xiaohong Cai
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
48
|
A HIF1a-Dependent Pro-Oxidant State Disrupts Synaptic Plasticity and Impairs Spatial Memory in Response to Intermittent Hypoxia. eNeuro 2020; 7:ENEURO.0024-20.2020. [PMID: 32493757 PMCID: PMC7363479 DOI: 10.1523/eneuro.0024-20.2020] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/30/2020] [Accepted: 05/15/2020] [Indexed: 02/06/2023] Open
Abstract
Sleep apnea causes cognitive deficits and is associated with several neurologic diseases. Intermittent hypoxia (IH) is recognized as a principal mediator of pathophysiology associated with sleep apnea, yet the basis by which IH contributes to impaired cognition remains poorly defined. Using a mouse model exposed to IH, this study examines how the transcription factor, hypoxia inducible factor 1a (HIF1a), contributes to disrupted synaptic physiology and spatial memory. In wild-type mice, impaired performance in the Barnes maze caused by IH coincided with a loss of NMDA receptor (NMDAr)-dependent long-term potentiation (LTP) in area CA1 and increased nuclear HIF1a within the hippocampus. IH-dependent HIF1a signaling caused a two-fold increase in expression of the reactive oxygen species (ROS) generating enzyme NADPH oxidase 4 (NOX4). These changes promoted a pro-oxidant state and the downregulation of GluN1 within the hippocampus. The IH-dependent effects were not present in either mice heterozygous for Hif1a (HIF1a+/-) or wild-type mice treated with the antioxidant manganese (III) tetrakis(1-methyl-4-pyridyl) porphyrin (MnTMPyP). Our findings indicate that HIF1a-dependent changes in redox state are central to the mechanism by which IH disrupts hippocampal synaptic plasticity and impairs spatial memory. This mechanism may enhance the vulnerability for cognitive deficit and lower the threshold for neurologic diseases associated untreated sleep apnea.
Collapse
|
49
|
Rubin BR, Milner TA, Pickel VM, Coleman CG, Marques-Lopes J, Van Kempen TA, Kazim SF, McEwen BS, Gray JD, Pereira AC. Sex and age differentially affect GABAergic neurons in the mouse prefrontal cortex and hippocampus following chronic intermittent hypoxia. Exp Neurol 2019; 325:113075. [PMID: 31837319 DOI: 10.1016/j.expneurol.2019.113075] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/22/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022]
Abstract
Obstructive sleep apnea (OSA), a chronic sleep disorder characterized by repetitive reduction or cessation of airflow during sleep, is widely prevalent and is associated with adverse neurocognitive sequelae including increased risk of Alzheimer's disease (AD). In humans, OSA is more common in elderly males. OSA is characterized by sleep fragmentation and chronic intermittent hypoxia (CIH), and recent epidemiological studies point to CIH as the best predictor of neurocognitive sequelae associated with OSA. The sex- and age- specific effects of OSA-associated CIH on specific cell populations such as γ-aminobutyric acid (GABA)-ergic neurons in the hippocampus and the medial prefrontal cortex (mPFC), regions important for cognitive function, remain largely unknown. The present study examined the effect of 35 days of either moderate (10% oxygen) or severe (5% oxygen) CIH on GABAergic neurons in the mPFC and hippocampus of young and aged male and female mice as well as post-accelerated ovarian failure (AOF) female mice. In the mPFC and hippocampus, the number of GABA-labeled neurons increased in aged and young severe CIH males compared to controls but not in young moderate CIH males. This change was not representative of the individual GABAergic cell subpopulations, as the number of parvalbumin-labeled neurons decreased while the number of somatostatin-labeled neurons increased in the hippocampus of severe CIH young males only. In all female groups, the number of GABA-labeled cells was not different between CIH and controls. However, in the mPFC, CIH increased the number of parvalbumin-labeled neurons in young females and the number of somatostatin-labeled cells in AOF females but decreased the number of somatostatin-labeled cells in aged females. In the hippocampus, CIH decreased the number of somatostatin-labeled neurons in young females. CIH decreased the density of vesicular GABA transporter in the mPFC of AOF females only. These findings suggest sex-specific changes in GABAergic neurons in the hippocampus and mPFC with males showing an increase of this cell population as compared to their female counterparts following CIH. Age at exposure and severity of CIH also differentially affect the GABAergic cell population in mice.
Collapse
Affiliation(s)
- Batsheva R Rubin
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States of America; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, United States of America.
| | - Teresa A Milner
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States of America; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, United States of America.
| | - Virginia M Pickel
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, United States of America
| | - Christal G Coleman
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, United States of America
| | - Jose Marques-Lopes
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, United States of America
| | - Tracey A Van Kempen
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, United States of America
| | - Syed Faraz Kazim
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States of America; Department of Neurology, Icahn School of Medicine, Mount Sinai, New York, NY 10029, United States of America; Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States of America
| | - Jason D Gray
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States of America
| | - Ana C Pereira
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States of America; Department of Neurology, Icahn School of Medicine, Mount Sinai, New York, NY 10029, United States of America; Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America.
| |
Collapse
|