51
|
Balan I, Bilger N, Saparov D, Hryb I, Abdyraimov A. Sleep Deprivation in Middle Age May Increase Dementia Risk: A Review. Cureus 2023; 15:e37425. [PMID: 37181993 PMCID: PMC10174673 DOI: 10.7759/cureus.37425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2023] [Indexed: 05/16/2023] Open
Abstract
Neurodegenerative diseases present increasing interest in clinical practice for the aging population and involve dysregulation of sleep-wake behaviors. Approximately 5.8 million adults aged 65 and older were living with Alzheimer's disease (AD) in the United States in 2020 with increased mortality compared to the declining cardiovascular and cancer death rates. We conducted an extensive literature review to evaluate and synthesize evidence regarding the association between short sleep duration or sleep deprivation and the risk of developing all-cause dementia and Alzheimer's disease. There are multiple mechanisms describing brain damage, such as brain hypoxia, oxidative stress, or blood-brain barrier (BBB) impairment, induced by chronic sleep restriction (CSR) and the potential correlation with future cognitive decline and dementia. More studies are necessary to identify the specific factors involved in the sleep loss-cognitive decline association that could be taken into consideration while elaborating recommendations for dementia prevention measures.
Collapse
Affiliation(s)
- Irina Balan
- Geriatrics, Montefiore Medical Center, Wakefield Campus, Bronx, USA
| | - Nataliya Bilger
- Clinical Simulation Center, Penn State University College of Medicine, Milton S. Hershey Medical Center, Hershey, USA
| | - Dosbai Saparov
- Internal Medicine, Brookdale University Hospital Medical Center, Brooklyn, USA
| | - Ihor Hryb
- Neuroscience, University of Minnesota, Minneapolis, USA
| | - Azamat Abdyraimov
- Biostatistics and Epidemiology, Ala-Too International University, Bishkek, KGZ
| |
Collapse
|
52
|
Sun Y, Zabihi M, Li Q, Li X, Kim BJ, Ubogu EE, Raja SN, Wesselmann U, Zhao C. Drug Permeability: From the Blood-Brain Barrier to the Peripheral Nerve Barriers. ADVANCED THERAPEUTICS 2023; 6:2200150. [PMID: 37649593 PMCID: PMC10465108 DOI: 10.1002/adtp.202200150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Indexed: 01/20/2023]
Abstract
Drug delivery into the peripheral nerves and nerve roots has important implications for effective local anesthesia and treatment of peripheral neuropathies and chronic neuropathic pain. Similar to drugs that need to cross the blood-brain barrier (BBB) and blood-spinal cord barrier (BSCB) to gain access to the central nervous system (CNS), drugs must cross the peripheral nerve barriers (PNB), formed by the perineurium and blood-nerve barrier (BNB) to modulate peripheral axons. Despite significant progress made to develop effective strategies to enhance BBB permeability in therapeutic drug design, efforts to enhance drug permeability and retention in peripheral nerves and nerve roots are relatively understudied. Guided by knowledge describing structural, molecular and functional similarities between restrictive neural barriers in the CNS and peripheral nervous system (PNS), we hypothesize that certain CNS drug delivery strategies are adaptable for peripheral nerve drug delivery. In this review, we describe the molecular, structural and functional similarities and differences between the BBB and PNB, summarize and compare existing CNS and peripheral nerve drug delivery strategies, and discuss the potential application of selected CNS delivery strategies to improve efficacious drug entry for peripheral nerve disorders.
Collapse
Affiliation(s)
- Yifei Sun
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Mahmood Zabihi
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Qi Li
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Xiaosi Li
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Brandon J. Kim
- Department of Biological Sciences, The University of Alabama, Tuscaloosa AL 35487, USA
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham AL 35294, USA
- Center for Convergent Biosciences and Medicine, University of Alabama, Tuscaloosa AL 35487, USA
- Alabama Life Research Institute, University of Alabama, Tuscaloosa AL 35487, USA
| | - Eroboghene E. Ubogu
- Division of Neuromuscular Disease, Department of Neurology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Srinivasa N. Raja
- Division of Pain Medicine, Department of Anesthesiology & Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Ursula Wesselmann
- Department of Anesthesiology and Perioperative Medicine, Division of Pain Medicine, and Department of Neurology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Consortium for Neuroengineering and Brain-Computer Interfaces, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Chao Zhao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
- Center for Convergent Biosciences and Medicine, University of Alabama, Tuscaloosa AL 35487, USA
- Alabama Life Research Institute, University of Alabama, Tuscaloosa AL 35487, USA
| |
Collapse
|
53
|
Contreras EG, Klämbt C. The Drosophila blood-brain barrier emerges as a model for understanding human brain diseases. Neurobiol Dis 2023; 180:106071. [PMID: 36898613 DOI: 10.1016/j.nbd.2023.106071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/24/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
The accurate regulation of the microenvironment within the nervous system is one of the key features characterizing complex organisms. To this end, neural tissue has to be physically separated from circulation, but at the same time, mechanisms must be in place to allow controlled transport of nutrients and macromolecules into and out of the brain. These roles are executed by cells of the blood-brain barrier (BBB) found at the interface of circulation and neural tissue. BBB dysfunction is observed in several neurological diseases in human. Although this can be considered as a consequence of diseases, strong evidence supports the notion that BBB dysfunction can promote the progression of brain disorders. In this review, we compile the recent evidence describing the contribution of the Drosophila BBB to the further understanding of brain disease features in human patients. We discuss the function of the Drosophila BBB during infection and inflammation, drug clearance and addictions, sleep, chronic neurodegenerative disorders and epilepsy. In summary, this evidence suggests that the fruit fly, Drosophila melanogaster, can be successfully employed as a model to disentangle mechanisms underlying human diseases.
Collapse
Affiliation(s)
- Esteban G Contreras
- University of Münster, Institute of Neuro- and Behavioral Biology, Badestr. 9, Münster, Germany.
| | - Christian Klämbt
- University of Münster, Institute of Neuro- and Behavioral Biology, Badestr. 9, Münster, Germany.
| |
Collapse
|
54
|
Chung Y, Wu JL, Huang WC. Effects of prebiotics on intestinal physiology, neuropsychological function, and exercise capacity of mice with sleep deprivation. Food Res Int 2023; 165:112568. [PMID: 36869551 DOI: 10.1016/j.foodres.2023.112568] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/27/2022] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
People suffered from insufficient or disrupted sleep due to night shifts, work pressure, and irregular lifestyles. Sleep deprivation caused by inadequate quantity or quality of sleep has been associated with not only increased risk of metabolic diseases, gut dysbiosis, and emotional disorders but also decreased work and exercise performance. In this study, we used the modified multiple platform method (MMPM) to induce pathological and psychological characteristics of sleep deprivation with C57BL/6J male mice, and investigated whether supplementing a prebiotics mixture of short-chain galactooligosaccharides (scGOS) and long-chain fructooligosaccharides (lcFOS) (9:1 ratio) could improve the impacts of sleep deprivation on intestinal physiology, neuropsychological function, inflammation, circadian rhythm, and exercise capacity. Results showed that sleep deprivation caused intestinal inflammation (increased TNFA and IL1B) and decreased intestinal permeability with a significant decrease in the tight junction genes (OCLN, CLDN1, TJP1, and TJP2) of intestine and brain. The prebiotics significantly increased the content of metabolite short-chain fatty acids (acetate and butyrate) while recovering the expression of indicated tight junction genes. In hypothalamus and hippocampus, clock (BMAL1 and CLOCK) and tight junction (OCLN and TJP2) genes were improved by prebiotics, and corticotropin-releasing hormone receptor genes, CRF1 and CRF2, were also significantly regulated for mitigation of depression and anxiety caused by sleep deprivation. Also, prebiotics brought significant benefits on blood sugar homeostasis and improvement of exercise performance. Functional prebiotics could improve physiological modulation, neuropsychological behaviors, and exercise performance caused by sleep deprivation, possibly through regulation of inflammation and circadian rhythm for health maintenance. However, the microbiota affected by prebiotics and sleep deprivation should warrant further investigation.
Collapse
Affiliation(s)
- Yi Chung
- College of Human Development and Health, National Taipei University of Nursing and Health Sciences, Taipei 112303, Taiwan.
| | - Jia-Ling Wu
- Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei 112303, Taiwan.
| | - Wen-Ching Huang
- Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei 112303, Taiwan.
| |
Collapse
|
55
|
Salminen A. Activation of aryl hydrocarbon receptor (AhR) in Alzheimer's disease: role of tryptophan metabolites generated by gut host-microbiota. J Mol Med (Berl) 2023; 101:201-222. [PMID: 36757399 PMCID: PMC10036442 DOI: 10.1007/s00109-023-02289-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/19/2022] [Accepted: 01/17/2023] [Indexed: 02/10/2023]
Abstract
Gut microbiota in interaction with intestinal host tissues influences many brain functions and microbial dysbiosis has been linked with brain disorders, such as neuropsychiatric conditions and Alzheimer's disease (AD). L-tryptophan metabolites and short-chained fatty acids (SCFA) are major messengers in the microbiota-brain axis. Aryl hydrocarbon receptors (AhR) are main targets of tryptophan metabolites in brain microvessels which possess an enriched expression of AhR protein. The Ah receptor is an evolutionarily conserved, ligand-activated transcription factor which is not only a sensor of xenobiotic toxins but also a pleiotropic regulator of both developmental processes and age-related tissue degeneration. Major microbiota-produced tryptophan metabolites involve indole derivatives, e.g., indole 3-pyruvic acid, indole 3-acetaldehyde, and indoxyl sulfate, whereas indoleamine and tryptophan 2,3-dioxygenases (IDO/TDO) of intestine host cells activate the kynurenine (KYN) pathway generating KYN metabolites, many of which are activators of AhR signaling. Chronic kidney disease (CKD) increases the serum level of indoxyl sulfate which promotes AD pathogenesis, e.g., it disrupts integrity of blood-brain barrier (BBB) and impairs cognitive functions. Activation of AhR signaling disturbs vascular homeostasis in brain; (i) it controls blood flow via the renin-angiotensin system, (ii) it inactivates endothelial nitric oxide synthase (eNOS), thus impairing NO production and vasodilatation, and (iii) it induces oxidative stress, stimulates inflammation, promotes cellular senescence, and enhances calcification of vascular walls. All these alterations are evident in cerebral amyloid angiopathy (CAA) in AD pathology. Moreover, AhR signaling can disturb circadian regulation and probably affect glymphatic flow. It seems plausible that dysbiosis of gut microbiota impairs the integrity of BBB via the activation of AhR signaling and thus aggravates AD pathology. KEY MESSAGES: Dysbiosis of gut microbiota is associated with dementia and Alzheimer's disease. Tryptophan metabolites are major messengers from the gut host-microbiota to brain. Tryptophan metabolites activate aryl hydrocarbon receptor (AhR) signaling in brain. The expression of AhR protein is enriched in brain microvessels and blood-brain barrier. Tryptophan metabolites disturb brain vascular integrity via AhR signaling. Dysbiosis of gut microbiota promotes inflammation and AD pathology via AhR signaling.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, Kuopio, 70211, Finland.
| |
Collapse
|
56
|
Dubayle D, Vanden-Bossche A, Peixoto T, Morel JL. Hypergravity Increases Blood-Brain Barrier Permeability to Fluorescent Dextran and Antisense Oligonucleotide in Mice. Cells 2023; 12:cells12050734. [PMID: 36899870 PMCID: PMC10000817 DOI: 10.3390/cells12050734] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 03/03/2023] Open
Abstract
The earliest effect of spaceflight is an alteration in vestibular function due to microgravity. Hypergravity exposure induced by centrifugation is also able to provoke motion sickness. The blood-brain barrier (BBB) is the crucial interface between the vascular system and the brain to ensure efficient neuronal activity. We developed experimental protocols of hypergravity on C57Bl/6JRJ mice to induce motion sickness and reveal its effects on the BBB. Mice were centrifuged at 2× g for 24 h. Fluorescent dextrans with different sizes (40, 70 and 150 kDa) and fluorescent antisense oligonucleotides (AS) were injected into mice retro-orbitally. The presence of fluorescent molecules was revealed by epifluorescence and confocal microscopies in brain slices. Gene expression was evaluated by RT-qPCR from brain extracts. Only the 70 kDa dextran and AS were detected in the parenchyma of several brain regions, suggesting an alteration in the BBB. Moreover, Ctnnd1, Gja4 and Actn1 were upregulated, whereas Jup, Tjp2, Gja1, Actn2, Actn4, Cdh2 and Ocln genes were downregulated, specifically suggesting a dysregulation in the tight junctions of endothelial cells forming the BBB. Our results confirm the alteration in the BBB after a short period of hypergravity exposure.
Collapse
Affiliation(s)
- David Dubayle
- CNRS, INCC, UMR 8002, Université Paris Cité, F-75006 Paris, France
- Correspondence: (D.D.); (J.-L.M.)
| | - Arnaud Vanden-Bossche
- INSERM, SAINBIOSE U1059, Mines Saint-Etienne, Université Jean Monnet Saint-Étienne, F-42023 Saint-Étienne, France
| | - Tom Peixoto
- University Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France
| | - Jean-Luc Morel
- University Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France
- University Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
- Correspondence: (D.D.); (J.-L.M.)
| |
Collapse
|
57
|
Segu A, Kannan NN. The duration of caffeine treatment plays an essential role in its effect on sleep and circadian rhythm. SLEEP ADVANCES : A JOURNAL OF THE SLEEP RESEARCH SOCIETY 2023; 4:zpad014. [PMID: 37193284 PMCID: PMC10108652 DOI: 10.1093/sleepadvances/zpad014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/28/2023] [Indexed: 05/18/2023]
Abstract
Sleep is regulated by the homeostatic system and the circadian clock. Caffeine intake promotes wakefulness in Drosophila. In humans, caffeine is consumed on a daily basis and hence it is important to understand the effect of prolonged caffeine intake on both circadian and homeostatic regulation of sleep. Furthermore, sleep changes with age and the impact of caffeine on age-dependent sleep fragmentation are yet to be understood. Hence in the present study, we examined the effect of short exposure to caffeine on homeostatic sleep and age-dependent sleep fragmentation in Drosophila. We further assessed the effect of prolonged exposure to caffeine on homeostatic sleep and circadian clock. The results of our study showed that short exposure to caffeine reduces sleep and food intake in mature flies. It also enhances sleep fragmentation with increasing age. However, we have not assessed the effect of caffeine on food intake in older flies. On the other hand, prolonged caffeine exposure did not exert any significant effect on the duration of sleep and food intake in mature flies. Nevertheless, prolonged caffeine ingestion decreased the morning and evening anticipatory activity in these flies indicating that it affects the circadian rhythm. These flies also exhibited phase delay in the clock gene timeless transcript oscillation and exhibited either behavioral arrhythmicity or a longer free-running period under constant darkness. In summary, the results of our studies showed that short exposure to caffeine increases the sleep fragmentation with age whereas prolonged caffeine exposure disrupts the circadian clock.
Collapse
Affiliation(s)
- Aishwarya Segu
- Chronobiology Laboratory, School of Biology, Indian Institute of Science Education and Research (IISER), Thiruvananthapuram, India
| | - Nisha N Kannan
- Chronobiology Laboratory, School of Biology, Indian Institute of Science Education and Research (IISER), Thiruvananthapuram, India
| |
Collapse
|
58
|
Whole CNS 3D Cryo-Fluorescence Tomography Shows CSF Clearance along Nasal Lymphatics, Spinal Nerves, and Lumbar/Sacral Lymph Nodes. J Imaging 2023; 9:jimaging9020045. [PMID: 36826964 PMCID: PMC9960470 DOI: 10.3390/jimaging9020045] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Unwanted proteins and metabolic waste in cerebral spinal fluid are cleared from the brain by meningeal and nasal lymphatics and the perineural sheath of cranial nerves; however, the distribution and clearance of cerebral spinal fluid (CSF) along the subarachnoid space of the entire spinal cord is not fully understood. Cryo-fluorescence tomography (CFT) was used to follow the movement of tracers from the ventricular system of the brain down through the meningeal lining of the spinal cord and out to the spinal lymphatic nodes. Isoflurane-anesthetized mice were infused into the lateral cerebroventricle with 5.0 µL of quantum dots [QdotR 605 ITKTM amino (PEG)] over two mins. Mice were allowed to recover (ca 2-3 min) and remained awake and ambulatory for 5, 15, 30, 60, and 120 min after which they were euthanized, and the entire intact body was frozen at -80°. The entire mouse was sectioned, and white light and fluorescent images were captured after each slice to produce high resolution three-dimensional volumes. Tracer appeared throughout the ventricular system and central canal of the spinal cord and the entire subarachnoid space of the CNS. A signal could be visualized in the nasal cavity, deep cervical lymph nodes, thoracic lymph nodes, and more superficial submandibular lymph nodes as early as 15 min post infusion. A fluorescent signal could be visualized along the dorsal root ganglia and down the proximal extension of the spinal nerves of the thoracic and lumbar segments at 30 min. There was a significant accumulation of tracer in the lumbar and sacral lymph nodes between 15-60 min. The dense fluorescent signal in the thoracic vertebrae noted at 5- and 15-min post infusion was significantly reduced by 30 min. Indeed, all signals in the spinal cord were ostensibly absent by 120 min, except for trace amounts in the coccyx. The brain still had some residual signal at 120 min. These data show that Qdots with a hydrodynamic diameter of 16-20 nm rapidly clear from the brain of awake mice. These data also clearly demonstrate the rapid distribution and efflux of traces along a major length of the vertebral column and the potential contribution of the spinal cord in the clearance of brain waste.
Collapse
|
59
|
Brain Waste Removal System and Sleep: Photobiomodulation as an Innovative Strategy for Night Therapy of Brain Diseases. Int J Mol Sci 2023; 24:ijms24043221. [PMID: 36834631 PMCID: PMC9965491 DOI: 10.3390/ijms24043221] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
Emerging evidence suggests that an important function of the sleeping brain is the removal of wastes and toxins from the central nervous system (CNS) due to the activation of the brain waste removal system (BWRS). The meningeal lymphatic vessels (MLVs) are an important part of the BWRS. A decrease in MLV function is associated with Alzheimer's and Parkinson's diseases, intracranial hemorrhages, brain tumors and trauma. Since the BWRS is activated during sleep, a new idea is now being actively discussed in the scientific community: night stimulation of the BWRS might be an innovative and promising strategy for neurorehabilitation medicine. This review highlights new trends in photobiomodulation of the BWRS/MLVs during deep sleep as a breakthrough technology for the effective removal of wastes and unnecessary compounds from the brain in order to increase the neuroprotection of the CNS as well as to prevent or delay various brain diseases.
Collapse
|
60
|
Schurhoff N, Toborek M. Circadian rhythms in the blood-brain barrier: impact on neurological disorders and stress responses. Mol Brain 2023; 16:5. [PMID: 36635730 PMCID: PMC9835375 DOI: 10.1186/s13041-023-00997-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 01/03/2023] [Indexed: 01/14/2023] Open
Abstract
Circadian disruption has become more prevalent in society due to the increase in shift work, sleep disruption, blue light exposure, and travel via different time zones. The circadian rhythm is a timed transcription-translation feedback loop with positive regulators, BMAL1 and CLOCK, that interact with negative regulators, CRY and PER, to regulate both the central and peripheral clocks. This review highlights the functions of the circadian rhythm, specifically in the blood-brain barrier (BBB), during both healthy and pathological states. The BBB is a highly selective dynamic interface composed of CNS endothelial cells, astrocytes, pericytes, neurons, and microglia that form the neurovascular unit (NVU). Circadian rhythms modulate BBB integrity through regulating oscillations of tight junction proteins, assisting in functions of the NVU, and modulating transporter functions. Circadian disruptions within the BBB have been observed in stress responses and several neurological disorders, including brain metastasis, epilepsy, Alzheimer's disease, and Parkinson's disease. Further understanding of these interactions may facilitate the development of improved treatment options and preventative measures.
Collapse
Affiliation(s)
- Nicolette Schurhoff
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Suite 528, 1011 NW 15th Street, Miami, FL, 33155, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Suite 528, 1011 NW 15th Street, Miami, FL, 33155, USA.
- Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, 40-065, Katowice, Poland.
| |
Collapse
|
61
|
Saunders NR, Dziegielewska KM, Fame RM, Lehtinen MK, Liddelow SA. The choroid plexus: a missing link in our understanding of brain development and function. Physiol Rev 2023; 103:919-956. [PMID: 36173801 PMCID: PMC9678431 DOI: 10.1152/physrev.00060.2021] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 09/01/2022] [Accepted: 09/17/2022] [Indexed: 11/22/2022] Open
Abstract
Studies of the choroid plexus lag behind those of the more widely known blood-brain barrier, despite a much longer history. This review has two overall aims. The first is to outline long-standing areas of research where there are unanswered questions, such as control of cerebrospinal fluid (CSF) secretion and blood flow. The second aim is to review research over the past 10 years where the focus has shifted to the idea that there are choroid plexuses located in each of the brain's ventricles that make specific contributions to brain development and function through molecules they generate for delivery via the CSF. These factors appear to be particularly important for aspects of normal brain growth. Most research carried out during the twentieth century dealt with the choroid plexus, a brain barrier interface making critical contributions to the composition and stability of the brain's internal environment throughout life. More recent research in the twenty-first century has shown the importance of choroid plexus-generated CSF in neurogenesis, influence of sex and other hormones on choroid plexus function, and choroid plexus involvement in circadian rhythms and sleep. The advancement of technologies to facilitate delivery of brain-specific therapies via the CSF to treat neurological disorders is a rapidly growing area of research. Conversely, understanding the basic mechanisms and implications of how maternal drug exposure during pregnancy impacts the developing brain represents another key area of research.
Collapse
Affiliation(s)
- Norman R Saunders
- Department of Neuroscience, The Alfred Centre, Monash University, Melbourne, Victoria, Australia
| | | | - Ryann M Fame
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, New York
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, New York
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, New York
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, New York
| |
Collapse
|
62
|
Calvo-Schimmel A, Kober KM, Paul SM, Cooper BA, Harris C, Shin J, Hammer MJ, Conley YP, Dokiparthi V, Olshen A, Levine JD, Miaskowski C. Sleep disturbance is associated with perturbations in immune-inflammatory pathways in oncology outpatients undergoing chemotherapy. Sleep Med 2023; 101:305-315. [PMID: 36470166 PMCID: PMC11200329 DOI: 10.1016/j.sleep.2022.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/14/2022] [Indexed: 11/23/2022]
Abstract
OBJECTIVE/BACKGROUND Sleep disturbance is a common problem in patients receiving chemotherapy. Purpose was to evaluate for perturbations in immune-inflammatory pathways between oncology patients with low versus very high levels of sleep disturbance. PATIENTS/METHODS Sleep disturbance was evaluated using the General Sleep Disturbance Scale six times over two cycles of chemotherapy. Latent profile analysis was used to identify subgroups of patients with distinct sleep disturbance profiles. Pathway impact analyses were performed in two independent samples using gene expression data obtained from RNA sequencing (n = 198) and microarray (n = 162) technologies. Fisher's combined probability test was used to identify significantly perturbed pathways between Low versus Very High sleep disturbance classes. RESULTS In the RNA sequencing and microarray samples, 59.1% and 51.9% of patients were in the Very High sleep disturbance class, respectively. Thirteen perturbed pathways were related to immune-inflammatory mechanisms (i.e., endocytosis, phagosome, antigen processing and presentation, natural killer cell mediated cytotoxicity, cytokine-cytokine receptor interaction, apoptosis, neutrophil extracellular trap formation, nucleotide-binding and oligomerization domain-like receptor signaling, Th17 cell differentiation, intestinal immune network for immunoglobulin A production, T-cell receptor signaling, complement and coagulation cascades, and tumor necrosis factor signaling). CONCLUSIONS First study to identify perturbations in immune-inflammatory pathways associated with very high levels of sleep disturbance in oncology outpatients. Findings suggest that complex immune-inflammatory interactions underlie sleep disturbance.
Collapse
Affiliation(s)
- Alejandra Calvo-Schimmel
- Department of Physiological Nursing, University of California, San Francisco, San Francisco, CA, USA.
| | - Kord M Kober
- Department of Physiological Nursing, University of California, San Francisco, San Francisco, CA, USA.
| | - Steven M Paul
- Department of Physiological Nursing, University of California, San Francisco, San Francisco, CA, USA.
| | - Bruce A Cooper
- Department of Physiological Nursing, University of California, San Francisco, San Francisco, CA, USA.
| | - Carolyn Harris
- Department of Physiological Nursing, University of California, San Francisco, San Francisco, CA, USA.
| | - Joosun Shin
- Department of Physiological Nursing, University of California, San Francisco, San Francisco, CA, USA.
| | | | - Yvette P Conley
- School of Nursing, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Vasuda Dokiparthi
- Department of Physiological Nursing, University of California, San Francisco, San Francisco, CA, USA.
| | - Adam Olshen
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA.
| | - Jon D Levine
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
| | - Christine Miaskowski
- Department of Physiological Nursing, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
63
|
Zhang Y, Zhao X, Zhang Y, Zeng F, Yan S, Chen Y, Li Z, Zhou D, Liu L. The role of circadian clock in astrocytes: From cellular functions to ischemic stroke therapeutic targets. Front Neurosci 2022; 16:1013027. [PMID: 36570843 PMCID: PMC9772621 DOI: 10.3389/fnins.2022.1013027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 11/10/2022] [Indexed: 12/14/2022] Open
Abstract
Accumulating evidence suggests that astrocytes, the abundant cell type in the central nervous system (CNS), play a critical role in maintaining the immune response after cerebral infarction, regulating the blood-brain barrier (BBB), providing nutrients to the neurons, and reuptake of glutamate. The circadian clock is an endogenous timing system that controls and optimizes biological processes. The central circadian clock and the peripheral clock are consistent, controlled by various circadian components, and participate in the pathophysiological process of astrocytes. Existing evidence shows that circadian rhythm controls the regulation of inflammatory responses by astrocytes in ischemic stroke (IS), regulates the repair of the BBB, and plays an essential role in a series of pathological processes such as neurotoxicity and neuroprotection. In this review, we highlight the importance of astrocytes in IS and discuss the potential role of the circadian clock in influencing astrocyte pathophysiology. A comprehensive understanding of the ability of the circadian clock to regulate astrocytes after stroke will improve our ability to predict the targets and biological functions of the circadian clock and gain insight into the basis of its intervention mechanism.
Collapse
Affiliation(s)
- Yuxing Zhang
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xin Zhao
- The Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ying Zhang
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Fukang Zeng
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Siyang Yan
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yao Chen
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhong Li
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Desheng Zhou
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,Desheng Zhou,
| | - Lijuan Liu
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,*Correspondence: Lijuan Liu,
| |
Collapse
|
64
|
Klymenko A, Lutz D. Melatonin signalling in Schwann cells during neuroregeneration. Front Cell Dev Biol 2022; 10:999322. [PMID: 36299487 PMCID: PMC9589221 DOI: 10.3389/fcell.2022.999322] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
It has widely been thought that in the process of nerve regeneration Schwann cells populate the injury site with myelinating, non–myelinating, phagocytic, repair, and mesenchyme–like phenotypes. It is now clear that the Schwann cells modify their shape and basal lamina as to accommodate re–growing axons, at the same time clear myelin debris generated upon injury, and regulate expression of extracellular matrix proteins at and around the lesion site. Such a remarkable plasticity may follow an intrinsic functional rhythm or a systemic circadian clock matching the demands of accurate timing and precision of signalling cascades in the regenerating nervous system. Schwann cells react to changes in the external circadian clock clues and to the Zeitgeber hormone melatonin by altering their plasticity. This raises the question of whether melatonin regulates Schwann cell activity during neurorepair and if circadian control and rhythmicity of Schwann cell functions are vital aspects of neuroregeneration. Here, we have focused on different schools of thought and emerging concepts of melatonin–mediated signalling in Schwann cells underlying peripheral nerve regeneration and discuss circadian rhythmicity as a possible component of neurorepair.
Collapse
|
65
|
Shi W, Chen C, Cui Q, Deng F, Yang B, Cao Y, Zhao F, Zhang Y, Du P, Wang J, Li T, Tang S, Shi X. Sleep disturbance exacerbates the cardiac conduction abnormalities induced by persistent heavy ambient fine particulate matter pollution: A multi-center cross-sectional study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 838:156472. [PMID: 35660605 DOI: 10.1016/j.scitotenv.2022.156472] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 06/15/2023]
Abstract
Fine particulate matter (PM2.5) exposure and sleep disturbance have been significantly associated with adverse cardiovascular outcomes, however, the combined effects of these two factors are still unclear. We conducted a multi-center cross-sectional study from November 2018 to May 2019 in the Beijing-Tianjin-Hebei region in China to investigate the potential modifying effects of sleep disturbance on associations between cardiac conduction abnormalities and PM2.5 exposure, as well as the combined effects of sleep disturbance and heavy pollution episodes, which were defined based on the PM2.5 mass concentration (≥75 μg/m3, falling in the 75th/90th percentile) and duration (1 day and ≥2 days). The sleep quality and sleep duration of all participants were evaluated using the Pittsburgh Sleep Quality Index. Standard 12-lead electrocardiogram (ECG) test was performed to measure the heart rate (HR), QRS duration (time taken for ventricular depolarization), HR corrected QT interval (time for ventricular depolarization and repolarization) and PR interval (time for atrioventricular conduction). Multivariable linear regression models were performed to evaluate the associations of PM2.5 and heavy pollution events on ECG parameters and the joint effects with sleep disturbance. We found PM2.5 exposure was independently associated with prolonged QRS and QTc intervals. Association between PM2.5 and the QTc interval was significantly stronger in participants with poor sleep quality. For each 10-μg/m3 increase in PM2.5 concentration, the QTc interval in the participants with poor sleep quality increased by 0.41 % (95 % confidence interval: 0.19, 0.64). In addition, heavy PM2.5 pollution episodes, especially extremely heavy pollution of long duration, were found to have synergistic effects with sleep disturbance on ECG parameters. Our findings provide evidence that PM2.5 exposure, especially heavy pollution episodes, may increase abnormal cardiac conduction and have a synergistic effect with sleep disturbance. Improving sleep hygiene is crucial to protect the heart health of the general population.
Collapse
Affiliation(s)
- Wanying Shi
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chen Chen
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qian Cui
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; School of Ecology and Environment, Inner Mongolia University, Hohhot, China
| | - Fuchang Deng
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Bo Yang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; School of Public Health, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Yaqiang Cao
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Feng Zhao
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yi Zhang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Peng Du
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jiaonan Wang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tiantian Li
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Song Tang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoming Shi
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
66
|
Liu X, Chiu JC. Nutrient-sensitive protein O-GlcNAcylation shapes daily biological rhythms. Open Biol 2022; 12:220215. [PMID: 36099933 PMCID: PMC9470261 DOI: 10.1098/rsob.220215] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/17/2022] [Indexed: 11/12/2022] Open
Abstract
O-linked-N-acetylglucosaminylation (O-GlcNAcylation) is a nutrient-sensitive protein modification that alters the structure and function of a wide range of proteins involved in diverse cellular processes. Similar to phosphorylation, another protein modification that targets serine and threonine residues, O-GlcNAcylation occupancy on cellular proteins exhibits daily rhythmicity and has been shown to play critical roles in regulating daily rhythms in biology by modifying circadian clock proteins and downstream effectors. We recently reported that daily rhythm in global O-GlcNAcylation observed in Drosophila tissues is regulated via the integration of circadian and metabolic signals. Significantly, mistimed feeding, which disrupts coordination of these signals, is sufficient to dampen daily O-GlcNAcylation rhythm and is predicted to negatively impact animal biological rhythms and health span. In this review, we provide an overview of published and potential mechanisms by which metabolic and circadian signals regulate hexosamine biosynthetic pathway metabolites and enzymes, as well as O-GlcNAc processing enzymes to shape daily O-GlcNAcylation rhythms. We also discuss the significance of functional interactions between O-GlcNAcylation and other post-translational modifications in regulating biological rhythms. Finally, we highlight organ/tissue-specific cellular processes and molecular pathways that could be modulated by rhythmic O-GlcNAcylation to regulate time-of-day-specific biology.
Collapse
Affiliation(s)
- Xianhui Liu
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California Davis, Davis, CA, USA
- Department of Pharmacology, School of Medicine, University of California Davis, Davis, CA, USA
| | - Joanna C. Chiu
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California Davis, Davis, CA, USA
| |
Collapse
|
67
|
Chichlowski M, Cotter J, Fawkes N, Pandey N. Feed your microbiome and improve sleep, stress resilience, and cognition. EXPLORATION OF MEDICINE 2022. [DOI: 10.37349/emed.2022.00097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The brain and gut are connected both physically and biochemically. The gut-brain axis includes the central nervous system, neuroendocrine and neuroimmune systems, the enteric nervous system and vagus nerve, and the gut microbiome. It can influence brain function and even behavior, suggesting that dietary interventions may help enhance and protect mental health and cognitive performance. This review focuses on the role of the microbiome and its metabolites in sleep regulation, neurodegenerative disorders, mechanisms of stress, and mood. It also provides examples of nutritional interventions which can restore healthy gut microbiota and aid with risk reduction and management of many disorders related to mental and cognitive health. Evidence suggests a shift in the gut microbiota towards a balanced composition could be a target to maintain brain health, reduce stress and improve quality of life.
Collapse
Affiliation(s)
- Maciej Chichlowski
- Medical and Scientific Affairs, Reckitt/Mead Johnson Nutrition Institute, Evansville, IN 47712, USA
| | - Jack Cotter
- Medical and Scientific Affairs, Reckitt/Mead Johnson Nutrition Institute, SL1 3UH Slough, UK
| | - Neil Fawkes
- Medical and Scientific Affairs, Reckitt/Mead Johnson Nutrition Institute, SL1 3UH Slough, UK
| | - Neeraj Pandey
- Medical and Scientific Affairs, Reckitt/Mead Johnson Nutrition Institute, SL1 3UH Slough, UK
| |
Collapse
|
68
|
Mohi-Ud-Din R, Mir RH, Mir PA, Banday N, Shah AJ, Sawhney G, Bhat MM, Batiha GE, Pottoo FH, Pottoo FH. Dysfunction of ABC Transporters at the Surface of BBB: Potential Implications in Intractable Epilepsy and Applications of Nanotechnology Enabled Drug Delivery. Curr Drug Metab 2022; 23:735-756. [PMID: 35980054 DOI: 10.2174/1389200223666220817115003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/10/2022] [Accepted: 05/31/2022] [Indexed: 01/05/2023]
Abstract
Epilepsy is a chronic neurological disorder affecting 70 million people globally. One of the fascinating attributes of brain microvasculature is the (BBB), which controls a chain of distinct features that securely regulate the molecules, ions, and cells movement between the blood and the parenchyma. The barrier's integrity is of paramount importance and essential for maintaining brain homeostasis, as it offers both physical and chemical barriers to counter pathogens and xenobiotics. Dysfunction of various transporters in the (BBB), mainly ATP binding cassette (ABC), is considered to play a vital role in hampering the availability of antiepileptic drugs into the brain. ABC (ATP-binding cassette) transporters constitute a most diverse protein superfamily, which plays an essential part in various biological processes, including cell homeostasis, cell signaling, uptake of nutrients, and drug metabolism. Moreover, it plays a crucial role in neuroprotection by out-flowing various internal and external toxic substances from the interior of a cell, thus decreasing their buildup inside the cell. In humans, forty-eight ABC transporters have been acknowledged and categorized into subfamilies A to G based on their phylogenetic analysis. ABC subfamilies B, C, and G, impart a vital role at the BBB in guarding the brain against the entrance of various xenobiotic and their buildup. The illnesses of the central nervous system have received a lot of attention lately Owing to the existence of the BBB, the penetration effectiveness of most CNS medicines into the brain parenchyma is very limited (BBB). In the development of neurological therapies, BBB crossing for medication delivery to the CNS continues to be a major barrier. Nanomaterials with BBB cross ability have indeed been extensively developed for the treatment of CNS diseases due to their advantageous properties. This review will focus on multiple possible factors like inflammation, oxidative stress, uncontrolled recurrent seizures, and genetic polymorphisms that result in the deregulation of ABC transporters in epilepsy and nanotechnology-enabled delivery across BBB in epilepsy.
Collapse
Affiliation(s)
- Roohi Mohi-Ud-Din
- Department of General Medicine, Sher-I-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, Jammu & Kashmir, 190011, India.,Department of Pharmaceutical Sciences, School of Applied Sciences & Technology, University of Kashmir, Hazratbal, Srinagar-190006, Jammu & Kashmir, India
| | - Reyaz Hassan Mir
- Pharmaceutical Chemistry Division, Chandigarh College of Pharmacy, Landran, Punjab-140301, India.,Department of Pharmaceutical Sciences, Pharmaceutical Chemistry Division, University of Kashmir, Hazratbal, Srinagar-190006, Kashmir, India
| | - Prince Ahad Mir
- Department of Pharmaceutical Sciences, Khalsa College of Pharmacy, G.T. Road, Amritsar-143002, Punjab, India
| | - Nazia Banday
- Department of Pharmaceutical Sciences, School of Applied Sciences & Technology, University of Kashmir, Hazratbal, Srinagar-190006, Jammu & Kashmir, India
| | - Abdul Jalil Shah
- Department of Pharmaceutical Sciences, Pharmaceutical Chemistry Division, University of Kashmir, Hazratbal, Srinagar-190006, Kashmir, India
| | - Gifty Sawhney
- Inflammation Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-Tawi, Jammu 180001, India
| | - Mudasir Maqbool Bhat
- Department of Pharmaceutical Sciences, Pharmacy Practice Division, University of Kashmir, Hazratbal, Srinagar-190006, Jammu & Kashmir, India
| | - Gaber E Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira, Egypt
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, 31441, Dammam, Saudi Arabia
| |
Collapse
|
69
|
Matsuoka RL, Buck LD, Vajrala KP, Quick RE, Card OA. Historical and current perspectives on blood endothelial cell heterogeneity in the brain. Cell Mol Life Sci 2022; 79:372. [PMID: 35726097 PMCID: PMC9209386 DOI: 10.1007/s00018-022-04403-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/18/2022] [Accepted: 05/25/2022] [Indexed: 11/28/2022]
Abstract
Dynamic brain activity requires timely communications between the brain parenchyma and circulating blood. Brain-blood communication is facilitated by intricate networks of brain vasculature, which display striking heterogeneity in structure and function. This vascular cell heterogeneity in the brain is fundamental to mediating diverse brain functions and has long been recognized. However, the molecular basis of this biological phenomenon has only recently begun to be elucidated. Over the past century, various animal species and in vitro systems have contributed to the accumulation of our fundamental and phylogenetic knowledge about brain vasculature, collectively advancing this research field. Historically, dye tracer and microscopic observations have provided valuable insights into the anatomical and functional properties of vasculature across the brain, and these techniques remain an important approach. Additionally, recent advances in molecular genetics and omics technologies have revealed significant molecular heterogeneity within brain endothelial and perivascular cell types. The combination of these conventional and modern approaches has enabled us to identify phenotypic differences between healthy and abnormal conditions at the single-cell level. Accordingly, our understanding of brain vascular cell states during physiological, pathological, and aging processes has rapidly expanded. In this review, we summarize major historical advances and current knowledge on blood endothelial cell heterogeneity in the brain, and discuss important unsolved questions in the field.
Collapse
Affiliation(s)
- Ryota L Matsuoka
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA. .,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA.
| | - Luke D Buck
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Keerti P Vajrala
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA.,Kansas City University College of Osteopathic Medicine, Kansas City, MO 64106, USA
| | - Rachael E Quick
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Olivia A Card
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
| |
Collapse
|
70
|
Berisha A, Shutkind K, Borniger JC. Sleep Disruption and Cancer: Chicken or the Egg? Front Neurosci 2022; 16:856235. [PMID: 35663547 PMCID: PMC9160986 DOI: 10.3389/fnins.2022.856235] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
Sleep is a nearly ubiquitous phenomenon across the phylogenetic tree, highlighting its essential role in ensuring fitness across evolutionary time. Consequently, chronic disruption of the duration, timing, or structure of sleep can cause widespread problems in multiple physiological systems, including those that regulate energy balance, immune function, and cognitive capacity, among others. Many, if not all these systems, become altered throughout the course of cancer initiation, growth, metastatic spread, treatment, and recurrence. Recent work has demonstrated how changes in sleep influence the development of chronic diseases, including cancer, in both humans and animal models. A common finding is that for some cancers (e.g., breast), chronic disruption of sleep/wake states prior to disease onset is associated with an increased risk for cancer development. Additionally, sleep disruption after cancer initiation is often associated with worse outcomes. Recently, evidence suggesting that cancer itself can affect neuronal circuits controlling sleep and wakefulness has accumulated. Patients with cancer often report difficulty falling asleep, difficulty staying asleep, and severe fatigue, during and even years after treatment. In addition to the psychological stress associated with cancer, cancer itself may alter sleep homeostasis through changes to host physiology and via currently undefined mechanisms. Moreover, cancer treatments (e.g., chemotherapy, radiation, hormonal, and surgical) may further worsen sleep problems through complex biological processes yet to be fully understood. This results in a "chicken or the egg" phenomenon, where it is unclear whether sleep disruption promotes cancer or cancer reciprocally disrupts sleep. This review will discuss existing evidence for both hypotheses and present a framework through which the interactions between sleep and cancer can be dissociated and causally investigated.
Collapse
Affiliation(s)
- Adrian Berisha
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States
| | - Kyle Shutkind
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | | |
Collapse
|
71
|
Gonçalves de Andrade E, González Ibáñez F, Tremblay MÈ. Microglia as a Hub for Suicide Neuropathology: Future Investigation and Prevention Targets. Front Cell Neurosci 2022; 16:839396. [PMID: 35663424 PMCID: PMC9158339 DOI: 10.3389/fncel.2022.839396] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/22/2022] [Indexed: 12/27/2022] Open
Abstract
Suicide is a complex public health challenge associated worldwide with one death every 40 s. Research advances in the neuropathology of suicidal behaviors (SB) have defined discrete brain changes which may hold the key to suicide prevention. Physiological differences in microglia, the resident immune cells of the brain, are present in post-mortem tissue samples of individuals who died by suicide. Furthermore, microglia are mechanistically implicated in the outcomes of important risk factors for SB, including early-life adversity, stressful life events, and psychiatric disorders. SB risk factors result in inflammatory and oxidative stress activities which could converge to microglial synaptic remodeling affecting susceptibility or resistance to SB. To push further this perspective, in this Review we summarize current areas of opportunity that could untangle the functional participation of microglia in the context of suicide. Our discussion centers around microglial state diversity in respect to morphology, gene and protein expression, as well as function, depending on various factors, namely brain region, age, and sex.
Collapse
Affiliation(s)
- Elisa Gonçalves de Andrade
- Neuroscience Graduate Program, Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Fernando González Ibáñez
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- *Correspondence: Marie-Ève Tremblay,
| |
Collapse
|
72
|
Mathew S, Sivasubbu S. Long Non Coding RNA Based Regulation of Cerebrovascular Endothelium. Front Genet 2022; 13:834367. [PMID: 35495157 PMCID: PMC9043600 DOI: 10.3389/fgene.2022.834367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
The rapid and high throughput discovery of long non coding RNAs (lncRNAs) has far outstripped the functional annotation of these novel transcripts in their respective cellular contexts. The cells of the blood brain barrier (BBB), especially the cerebrovascular endothelial cells (CVECs), are strictly regulated to maintain a controlled state of homeostasis for undisrupted brain function. Several key pathways are understood in CVEC function that lead to the development and maintenance of their barrier properties, the dysregulation of which leads to BBB breakdown and neuronal injury. Endothelial lncRNAs have been discovered and functionally validated in the past decade, spanning a wide variety of regulatory mechanisms in health and disease. We summarize here the lncRNA-mediated regulation of established pathways that maintain or disrupt the barrier property of CVECs, including in conditions such as ischemic stroke and glioma. These lncRNAs namely regulate the tight junction assembly/disassembly, angiogenesis, autophagy, apoptosis, and so on. The identification of these lncRNAs suggests a less understood mechanistic layer, calling for further studies in appropriate models of the blood brain barrier to shed light on the lncRNA-mediated regulation of CVEC function. Finally, we gather various approaches for validating lncRNAs in BBB function in human organoids and animal models and discuss the therapeutic potential of CVEC lncRNAs along with the current limitations.
Collapse
Affiliation(s)
- Samatha Mathew
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sridhar Sivasubbu
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India,*Correspondence: Sridhar Sivasubbu,
| |
Collapse
|
73
|
Hetman M, Slomnicki L, Hodges E, Ohri SS, Whittemore SR. Role of circadian rhythms in pathogenesis of acute CNS injuries: Insights from experimental studies. Exp Neurol 2022; 353:114080. [DOI: 10.1016/j.expneurol.2022.114080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/28/2022] [Accepted: 04/05/2022] [Indexed: 11/16/2022]
|
74
|
Markowicz-Piasecka M, Markiewicz A, Darłak P, Sikora J, Adla SK, Bagina S, Huttunen KM. Current Chemical, Biological, and Physiological Views in the Development of Successful Brain-Targeted Pharmaceutics. Neurotherapeutics 2022; 19:942-976. [PMID: 35391662 PMCID: PMC9294128 DOI: 10.1007/s13311-022-01228-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2022] [Indexed: 12/13/2022] Open
Abstract
One of the greatest challenges with successful pharmaceutical treatments of central nervous system (CNS) diseases is the delivery of drugs into their target sites with appropriate concentrations. For example, the physically tight blood-brain barrier (BBB) effectively blocks compounds from penetrating into the brain, also by the action of metabolizing enzymes and efflux transport mechanisms. However, many endogenous compounds, including both smaller compounds and macromolecules, like amino acids, sugars, vitamins, nucleosides, hormones, steroids, and electrolytes, have their peculiar internalization routes across the BBB. These delivery mechanisms, namely carrier-mediated transport and receptor-mediated transcytosis have been utilized to some extent in brain-targeted drug development. The incomplete knowledge of the BBB and the smaller than a desirable number of chemical tools have hindered the development of successful brain-targeted pharmaceutics. This review discusses the recent advancements achieved in the field from the point of medicinal chemistry view and discusses how brain drug delivery can be improved in the future.
Collapse
Affiliation(s)
- Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego1, 90-151 Lodz, Poland
| | - Agata Markiewicz
- Students Research Group, Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland
| | - Patrycja Darłak
- Students Research Group, Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland
| | - Joanna Sikora
- Department of Bioinorganic Chemistry, Medical University of Lodz, Medical University of Lodz, ul. Muszyńskiego1, 90-151 Lodz, Poland
| | - Santosh Kumar Adla
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211 Kuopio, Finland
- Institute of Organic Chemistry and Biochemistry (IOCB), Czech Academy of Sciences, Flemingovo Namesti 542/2, 160 00 Prague, Czech Republic
| | - Sreelatha Bagina
- Charles River Discovery Research Services Finland Oy, Neulaniementie 4, 70210 Kuopio, Finland
| | - Kristiina M. Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211 Kuopio, Finland
| |
Collapse
|
75
|
Salehpour F, Khademi M, Bragin DE, DiDuro JO. Photobiomodulation Therapy and the Glymphatic System: Promising Applications for Augmenting the Brain Lymphatic Drainage System. Int J Mol Sci 2022; 23:ijms23062975. [PMID: 35328396 PMCID: PMC8950470 DOI: 10.3390/ijms23062975] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 12/21/2022] Open
Abstract
The glymphatic system is a glial-dependent waste clearance pathway in the central nervous system, devoted to drain away waste metabolic products and soluble proteins such as amyloid-beta. An impaired brain glymphatic system can increase the incidence of neurovascular, neuroinflammatory, and neurodegenerative diseases. Photobiomodulation (PBM) therapy can serve as a non-invasive neuroprotective strategy for maintaining and optimizing effective brain waste clearance. In this review, we discuss the crucial role of the glymphatic drainage system in removing toxins and waste metabolites from the brain. We review recent animal research on the neurotherapeutic benefits of PBM therapy on glymphatic drainage and clearance. We also highlight cellular mechanisms of PBM on the cerebral glymphatic system. Animal research has shed light on the beneficial effects of PBM on the cerebral drainage system through the clearance of amyloid-beta via meningeal lymphatic vessels. Finally, PBM-mediated increase in the blood–brain barrier permeability with a subsequent rise in Aβ clearance from PBM-induced relaxation of lymphatic vessels via a vasodilation process will be discussed. We conclude that PBM promotion of cranial and extracranial lymphatic system function might be a promising strategy for the treatment of brain diseases associated with cerebrospinal fluid outflow abnormality.
Collapse
Affiliation(s)
- Farzad Salehpour
- College for Light Medicine and Photobiomodulation, D-82319 Starnberg, Germany;
- ProNeuroLIGHT LLC, Phoenix, AZ 85041, USA
| | - Mahsa Khademi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz 51666, Iran;
| | - Denis E. Bragin
- Department of Neurology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA;
| | - Joseph O. DiDuro
- ProNeuroLIGHT LLC, Phoenix, AZ 85041, USA
- Correspondence: ; Tel.: +1-(845)-203-9204
| |
Collapse
|
76
|
McConnell HL, Mishra A. Cells of the Blood-Brain Barrier: An Overview of the Neurovascular Unit in Health and Disease. Methods Mol Biol 2022; 2492:3-24. [PMID: 35733036 PMCID: PMC9987262 DOI: 10.1007/978-1-0716-2289-6_1] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
The brain is endowed with highly specialized vasculature that is both structurally and functionally unique compared to vasculature supplying peripheral organs. The blood-brain barrier (BBB) is formed by endothelial cells of the cerebral vasculature and prevents extravasation of blood products into the brain to protect neural tissue and maintain a homeostatic environment. The BBB functions as part of the neurovascular unit (NVU), which is composed of neurons, astrocytes, and microglia in addition to the specialized endothelial cells, mural cells, and the basement membrane. Through coordinated intercellular signaling, these cells function as a dynamic unit to tightly regulate brain blood flow, vascular function, neuroimmune responses, and waste clearance. In this chapter, we review the functions of individual NVU components, describe neurovascular coupling as a classic example of NVU function, and discuss archetypal NVU pathophysiology during disease.
Collapse
Affiliation(s)
- Heather L McConnell
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, OR, USA
- Office of Academic Development, Houston Methodist Research Institute, Houston, TX, USA
| | - Anusha Mishra
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, OR, USA.
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
77
|
Contreras EG, Sierralta J. The Fly Blood-Brain Barrier Fights Against Nutritional Stress. Neurosci Insights 2022; 17:26331055221120252. [PMID: 36225749 PMCID: PMC9549514 DOI: 10.1177/26331055221120252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
In the wild, animals face different challenges including multiple events of food
scarcity. How they overcome these conditions is essential for survival. Thus,
adaptation mechanisms evolved to allow the development and survival of an
organism during nutrient restriction periods. Given the high energy demand of
the nervous system, the molecular mechanisms of adaptation to malnutrition are
of great relevance to fuel the brain. The blood-brain barrier (BBB) is the
interface between the central nervous system (CNS) and the circulatory system.
The BBB mediates the transport of macromolecules in and out of the CNS, and
therefore, it can buffer changes in nutrient availability. In this review, we
collect the current evidence using the fruit fly, Drosophila
melanogaster, as a model of the role of the BBB in the adaptation
to starvation. We discuss the role of the Drosophila BBB during
nutrient deprivation as a potential sensor for circulating nutrients, and
transient nutrient storage as a regulator of the CNS neurogenic niche.
Collapse
Affiliation(s)
- Esteban G Contreras
- Institute of Neuro- and Behavioral Biology, University of Münster, Münster, Germany
| | - Jimena Sierralta
- Biomedical Neuroscience Institute and Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
78
|
Multi-Modal Regulation of Circadian Physiology by Interactive Features of Biological Clocks. BIOLOGY 2021; 11:biology11010021. [PMID: 35053019 PMCID: PMC8772734 DOI: 10.3390/biology11010021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 12/26/2022]
Abstract
The circadian clock is a fundamental biological timing mechanism that generates nearly 24 h rhythms of physiology and behaviors, including sleep/wake cycles, hormone secretion, and metabolism. Evolutionarily, the endogenous clock is thought to confer living organisms, including humans, with survival benefits by adapting internal rhythms to the day and night cycles of the local environment. Mirroring the evolutionary fitness bestowed by the circadian clock, daily mismatches between the internal body clock and environmental cycles, such as irregular work (e.g., night shift work) and life schedules (e.g., jet lag, mistimed eating), have been recognized to increase the risk of cardiac, metabolic, and neurological diseases. Moreover, increasing numbers of studies with cellular and animal models have detected the presence of functional circadian oscillators at multiple levels, ranging from individual neurons and fibroblasts to brain and peripheral organs. These oscillators are tightly coupled to timely modulate cellular and bodily responses to physiological and metabolic cues. In this review, we will discuss the roles of central and peripheral clocks in physiology and diseases, highlighting the dynamic regulatory interactions between circadian timing systems and multiple metabolic factors.
Collapse
|
79
|
Li Y, Shao L, Mou Y, Zhang Y, Ping Y. Sleep, circadian rhythm and gut microbiota: alterations in Alzheimer's disease and their potential links in the pathogenesis. Gut Microbes 2021; 13:1957407. [PMID: 34520319 PMCID: PMC8463034 DOI: 10.1080/19490976.2021.1957407] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
ABSTRATCIn recent years, emerging studies have observed gut microbiota (GM) alterations in Alzheimer's disease (AD), even in individuals with mild cognitive impairment (MCI). Further, impaired sleep and circadian patterns are common symptoms of AD, while sleep and circadian rhythm disruption (SCRD) is associated with greater β-amyloid (Aβ) burden and AD risk, sometimes years before the clinical onset of AD. Moreover, reports have demonstrated that GM and its metabolites exhibit diurnal rhythmicity and the role of SCRD in dampening the GM rhythmicity and eubiosis. This review will provide an evaluation of clinical and animal studies describing GM alterations in distinct conditions, including AD, sleep and circadian disruption. It aims to identify the overlapping and distinctive GM alterations in these conditions and their contributions to pathophysiology. Although most studies are observational and use different methodologies, data indicate partial commonalities in GM alterations and unanimity at functional level. Finally, we discuss the possible interactions between SCRD and GM in AD pathogenesis, as well as several methodological improvements that are necessary for future research.
Collapse
Affiliation(s)
- Yi Li
- School of Medicine, Imperial College London, London, UK
| | - Lingzhan Shao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Yang Mou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Yan Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Yong Ping
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China,Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,CONTACT Yong Ping Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
80
|
Walker WH, Sprowls SA, Bumgarner JR, Liu JA, Meléndez-Fernández OH, Walton JC, Lockman PR, DeVries AC, Nelson RJ. Circadian Influences on Chemotherapy Efficacy in a Mouse Model of Brain Metastases of Breast Cancer. Front Oncol 2021; 11:752331. [PMID: 34956876 PMCID: PMC8695439 DOI: 10.3389/fonc.2021.752331] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/19/2021] [Indexed: 11/30/2022] Open
Abstract
Chemotherapy is more effective in the treatment of peripheral tumors than brain metastases, likely reflecting the reduced ability of chemotherapy to cross the blood-brain barrier (BBB) and blood-tumor barrier at efficacious concentrations. Recent studies demonstrate circadian regulation of the BBB. Thus, we predicted that optimally timed chemotherapy would increase anti-tumor efficacy in a model of brain metastases of breast cancer (BMBC). First, we characterized novel daily alterations in BBB permeability to a commonly used chemotherapeutic, 14C-paclitaxel, within BMBC following injections given at four time points across the day. Peak and trough 14C-paclitaxel concentrations within BMBC occurred during the mid-dark phase and at the beginning of the light phase, respectively. Notably, chemotherapy injections during the dark phase increased cell death within BMBC and delayed onset of neurological symptoms relative to injections during the light phase. These data provide strong evidence for the beneficial effects of chrono-chemotherapy for the treatment of BMBC.
Collapse
Affiliation(s)
- William H. Walker
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Samuel A. Sprowls
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, United States
| | - Jacob R. Bumgarner
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Jennifer A. Liu
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | | | - James C. Walton
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Paul R. Lockman
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, United States
- WVU Cancer Institute, West Virginia University, Morgantown, WV, United States
| | - A. Courtney DeVries
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
- WVU Cancer Institute, West Virginia University, Morgantown, WV, United States
- Department of Medicine, Division of Oncology/Hematology, West Virginia University, Morgantown, WV, United States
| | - Randy J. Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
81
|
Jaggard JB, Wang GX, Mourrain P. Non-REM and REM/paradoxical sleep dynamics across phylogeny. Curr Opin Neurobiol 2021; 71:44-51. [PMID: 34583217 PMCID: PMC8719594 DOI: 10.1016/j.conb.2021.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 12/14/2022]
Abstract
All animals carefully studied sleep, suggesting that sleep as a behavioral state exists in all animal life. Such evolutionary maintenance of an otherwise vulnerable period of environmental detachment suggests that sleep must be integral in fundamental biological needs. Despite over a century of research, the knowledge of what sleep does at the tissue, cellular or molecular levels remain cursory. Currently, sleep is defined based on behavioral criteria and physiological measures rather than at the cellular or molecular level. Physiologically, sleep has been described as two main states, non-rapid eye moment (NREM) and REM/paradoxical sleep (PS), which are defined in the neocortex by synchronous oscillations and paradoxical wake-like activity, respectively. For decades, these two sleep states were believed to be defining characteristics of only mammalian and avian sleep. Recent work has revealed slow oscillation, silencing, and paradoxical/REM-like activities in reptiles, fish, flies, worms, and cephalopods suggesting that these sleep dynamics and associated physiological states may have emerged early in animal evolution. Here, we discuss these recent developments supporting the conservation of neural dynamics (silencing, oscillation, paradoxical activity) of sleep states across phylogeny.
Collapse
Affiliation(s)
- James B Jaggard
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Gordon X Wang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Wu Tsai Neuroscience Institute, Stanford University, Stanford, CA, USA
| | - Philippe Mourrain
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; INSERM 1024, Ecole Normale Supérieure, Paris, France.
| |
Collapse
|
82
|
Garcia P, Montastruc JL, Rousseau V, Hamard J, Sommet A, Montastruc F. β-adrenoceptor antagonists and nightmares: A pharmacoepidemiological-pharmacodynamic study. J Psychopharmacol 2021; 35:1441-1448. [PMID: 34318729 DOI: 10.1177/02698811211034810] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AIM To compare different β-adrenoceptor antagonists for the risk of reporting nightmare. METHODS The study involved two approaches: first, we investigated in VigiBase®, the World Health Organization Individual Case Safety Report (ICSR) database, the disproportionality between exposure to each β-adrenoceptor antagonists and reports of nightmares between 1967 and 2019. Second, in a pharmacoepidemiological-pharmacodynamic analysis, we assessed whether use of β-adrenoceptor antagonists with moderate and high lipid solubility or strong 5-HT1A affinity were associated with an increased risk of reporting nightmares. We conducted multivariate logistic regression to estimate reporting odds ratios (RORs) of nightmares compared to all other adverse drug reactions. RESULTS Of the 126,964 reports recorded with β-adrenoceptor antagonists, 1138 (0.9%) were nightmares. The highest risk of reporting a nightmare was found with exposure of pindolol (adjusted ROR 2.82, 95%CI, 2.19-3.61), metoprolol (1.89, 1.66-2.16), and alprenolol (1.77, 1.06-2.97). Compared to use of low lipid solubility β-adrenoceptor antagonists, use of moderate or high lipid solubility β-adrenoceptor antagonists were significantly more associated with nightmare reports (aROR moderate vs. low 1.72, 95%CI 1.47-2.00 and aROR high vs. low 1.84, 95%CI 1.53-2.22). Use of moderate or high 5-HT1A affinity of β-adrenoceptor antagonists was associated with an increased ROR of nightmares compared with low 5-HT1A affinity of β-adrenoceptor antagonists (aROR moderate vs. low 1.22, 95%CI 1.04-1.43 and aROR high vs. low 2.46, 95%CI 1.93-3.13). CONCLUSION In our large pharmacovigilance study, nightmares are more frequently reported for pindolol and metoprolol, and among β-adrenoceptor antagonists with high lipid solubility and high 5-HT1A receptor affinity.
Collapse
Affiliation(s)
- Philippe Garcia
- Department of Medical and Clinical Pharmacology, Centre of Pharmacovigilance and Pharmacoepidemiology, Faculty of Medicine, Toulouse University Hospital, Toulouse, France
| | - Jean-Louis Montastruc
- Department of Medical and Clinical Pharmacology, Centre of Pharmacovigilance and Pharmacoepidemiology, Faculty of Medicine, Toulouse University Hospital, Toulouse, France.,CIC 1436, Team PEPSS-Pharmacologie En Population cohorteS et biobanqueS, Toulouse University Hospital, Toulouse, France
| | - Vanessa Rousseau
- Department of Medical and Clinical Pharmacology, Centre of Pharmacovigilance and Pharmacoepidemiology, Faculty of Medicine, Toulouse University Hospital, Toulouse, France.,CIC 1436, Team PEPSS-Pharmacologie En Population cohorteS et biobanqueS, Toulouse University Hospital, Toulouse, France
| | - Jacques Hamard
- Department of Medical and Clinical Pharmacology, Centre of Pharmacovigilance and Pharmacoepidemiology, Faculty of Medicine, Toulouse University Hospital, Toulouse, France
| | - Agnès Sommet
- Department of Medical and Clinical Pharmacology, Centre of Pharmacovigilance and Pharmacoepidemiology, Faculty of Medicine, Toulouse University Hospital, Toulouse, France.,CIC 1436, Team PEPSS-Pharmacologie En Population cohorteS et biobanqueS, Toulouse University Hospital, Toulouse, France
| | - François Montastruc
- Department of Medical and Clinical Pharmacology, Centre of Pharmacovigilance and Pharmacoepidemiology, Faculty of Medicine, Toulouse University Hospital, Toulouse, France.,CIC 1436, Team PEPSS-Pharmacologie En Population cohorteS et biobanqueS, Toulouse University Hospital, Toulouse, France
| |
Collapse
|
83
|
Slomnicki LP, Wei G, Burke DA, Hodges ER, Myers SA, Yarberry CD, Morehouse JR, Whittemore SR, Saraswat Ohri S, Hetman M. Limited changes in locomotor recovery and unaffected white matter sparing after spinal cord contusion at different times of day. PLoS One 2021; 16:e0249981. [PMID: 34813603 PMCID: PMC8610253 DOI: 10.1371/journal.pone.0249981] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 10/20/2021] [Indexed: 11/18/2022] Open
Abstract
The circadian gene expression rhythmicity drives diurnal oscillations of physiological processes that may determine the injury response. While outcomes of various acute injuries are affected by the time of day at which the original insult occurred, such influences on recovery after spinal cord injury (SCI) are unknown. We report that mice receiving moderate, T9 contusive SCI at ZT0 (zeitgeber time 0, time of lights on) and ZT12 (time of lights off) showed similar hindlimb function recovery in the Basso mouse scale (BMS) over a 6 week post-injury period. In an independent study, no significant differences in BMS were observed after SCI at ZT18 vs. ZT6. However, the ladder walking test revealed modestly improved performance for ZT18 vs. ZT6 mice at week 6 after injury. Consistent with those minor effects on functional recovery, terminal histological analysis revealed no significant differences in white matter sparing at the injury epicenter. Likewise, blood-spinal cord barrier disruption and neuroinflammation appeared similar when analyzed at 1 week post injury at ZT6 or ZT18. Therefore, locomotor recovery after thoracic contusive SCI is not substantively modulated by the time of day at which the neurotrauma occurred.
Collapse
Affiliation(s)
- Lukasz P. Slomnicki
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - George Wei
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Darlene A. Burke
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Emily R. Hodges
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Scott A. Myers
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Christine D. Yarberry
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Johnny R. Morehouse
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Scott R. Whittemore
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Department of Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Michal Hetman
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Department of Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
84
|
Neal EH, Katdare KA, Shi Y, Marinelli NA, Hagerla KA, Lippmann ES. Influence of basal media composition on barrier fidelity within human pluripotent stem cell-derived blood-brain barrier models. J Neurochem 2021; 159:980-991. [PMID: 34716922 DOI: 10.1111/jnc.15532] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 10/03/2021] [Accepted: 10/15/2021] [Indexed: 01/02/2023]
Abstract
It is increasingly recognized that brain microvascular endothelial cells (BMECs), the principal component of the blood-brain barrier (BBB), are highly sensitive to soluble cues from both the bloodstream and the brain. This concept extends in vitro, where the extracellular milieu can also influence BBB properties in cultured cells. However, the extent to which baseline culture conditions can affect BBB properties in vitro remains unclear, which has implications for model variability and reproducibility, as well as downstream assessments of molecular transport and disease phenotypes. Here, we explore this concept by examining BBB properties within human-induced pluripotent stem cell (iPSC)-derived BMEC-like cells cultured under serum-free conditions in DMEM/F12 and Neurobasal media, which have fully defined compositions. We demonstrate notable differences in both passive and active BBB properties as a function of basal media composition. Further, RNA sequencing and phosphoproteome analyses revealed alterations to various signaling pathways in response to basal media differences. Overall, our results demonstrate that baseline culture conditions can have a profound influence on the performance of in vitro BBB models, and these effects should be considered when designing experiments that utilize such models for basic research and preclinical assays.
Collapse
Affiliation(s)
- Emma H Neal
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Ketaki A Katdare
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - Yajuan Shi
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Nicholas A Marinelli
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, Tennessee, USA
| | - Kameron A Hagerla
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - Ethan S Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA.,Chemical and Physical Biology Program, Vanderbilt University, Nashville, Tennessee, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA.,Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
85
|
Cuddapah VA, Goldberg EM. REVing up the Brain: A Mechanism Driving Seizure Timing. Epilepsy Curr 2021; 22:64-65. [PMID: 35233204 PMCID: PMC8832345 DOI: 10.1177/15357597211054257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
86
|
Marques CG, Dos Santos Quaresma MVL, Nakamoto FP, Magalhães ACO, Lucin GA, Thomatieli-Santos RV. Does Modern Lifestyle Favor Neuroimmunometabolic Changes? A Path to Obesity. Front Nutr 2021; 8:705545. [PMID: 34621773 PMCID: PMC8490681 DOI: 10.3389/fnut.2021.705545] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/27/2021] [Indexed: 12/19/2022] Open
Abstract
Factors linked to modern lifestyles, such as physical inactivity, Western diet, and poor sleep quality have been identified as key contributors to the positive energy balance (PEB). PEB rises adipose tissue hypertrophy and dysfunction over the years, affecting cells and tissues that are metabolically critical for energy homeostasis regulation, especially skeletal muscle, hypothalamic-pituitary-adrenal axis, and gut microbiota. It is known that the interaction among lifestyle factors and tissue metabolic dysfunction increases low-grade chronic systemic inflammation, leading to insulin resistance and other adverse metabolic disorders. Although immunometabolic mechanisms are widely discussed in obesity, neuroimmunoendocrine pathways have gained notoriety, as a link to neuroinflammation and central nervous system disorders. Hypothalamic inflammation has been associated with food intake dysregulation, which comprises homeostatic and non-homeostatic mechanisms, promoting eating behavior changes related to the obesity prevalence. The purpose of this review is to provide an updated and integrated perspective on the effects of Western diet, sleep debt, and physical exercise on the regulation of energy homeostasis and low-grade chronic systemic inflammation. Subsequently, we discuss the intersection between systemic inflammation and neuroinflammation and how it can contribute to energy imbalance, favoring obesity. Finally, we propose a model of interactions between systemic inflammation and neuroinflammation, providing new insights into preventive and therapeutic targets for obesity.
Collapse
Affiliation(s)
- Camila Guazzelli Marques
- Programa de Pós-graduação em Psicobiologia, Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - Ana Carolina Oumatu Magalhães
- Programa de Pós-graduação em Psicobiologia, Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil.,Departamento de Nutrição, Centro Universitário São Camilo, São Paulo, Brazil
| | | | - Ronaldo Vagner Thomatieli-Santos
- Programa de Pós-graduação em Psicobiologia, Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil.,Departamento de Biociências, Universidade Federal de São Paulo, Santos, Brazil
| |
Collapse
|
87
|
Tremblay MÈ. Microglial functional alteration and increased diversity in the challenged brain: Insights into novel targets for intervention. Brain Behav Immun Health 2021; 16:100301. [PMID: 34589793 PMCID: PMC8474548 DOI: 10.1016/j.bbih.2021.100301] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 02/07/2023] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS) parenchyma, which perform beneficial physiological roles across life. These immune cells actively maintain CNS health by clearing toxic debris and removing dysfunctional or degenerating cells. They also modify the wiring of neuronal circuits, by acting on the formation, modification, and elimination of synapses-the connections between neurons. Microglia furthermore recently emerged as highly diverse cells comprising several structural and functional states, indicating a far more critical involvement in orchestrating brain development, plasticity, behaviour, and cognition. Various environmental factors, together with the individual genetic predispositions, confer an increased risk for neurodevelopmental and neuropsychiatric disorders, as well as neurodegenerative diseases that include autism spectrum disorders, schizophrenia, major depressive disorder, and Alzheimer's disease, across life. Microglia are highly sensitive to chronic psychological stress, inadequate diet, viral/bacterial infection, pollution, and insufficient or altered sleep, especially during critical developmental periods, but also throughout life. These environmental challenges can compromise microglial physiological functions, resulting notably in defective neuronal circuit wiring, altered brain functional connectivity, and the onset of behavioral deficits into adolescence, adulthood, and aging. This short review provides a historical and technical perspective, notably focused on my contribution to the field, on how environmental challenges affect microglia, particularly their physiological functions, and increase their diversity, which provides novel targets for intervention.
Collapse
Affiliation(s)
- Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Molecular Medicine Department, Université Laval, Québec City, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- The Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
88
|
Lysen TS, Yilmaz P, Dubost F, Ikram MA, de Bruijne M, Vernooij MW, Luik AI. Sleep and perivascular spaces in the middle-aged and elderly population. J Sleep Res 2021; 31:e13485. [PMID: 34549850 PMCID: PMC9285071 DOI: 10.1111/jsr.13485] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/13/2021] [Accepted: 09/01/2021] [Indexed: 12/29/2022]
Abstract
Sleep has been hypothesised to facilitate waste clearance from the brain. We aimed to determine whether sleep is associated with perivascular spaces on brain magnetic resonance imaging (MRI), a potential marker of impaired brain waste clearance, in a population-based cohort of middle-aged and elderly people. In 559 participants (mean [SD] age 62 [6] years, 52% women) from the population-based Rotterdam Study, we measured total sleep time, sleep onset latency, wake after sleep onset and sleep efficiency with actigraphy and polysomnography. Perivascular space load was determined with brain MRI in four regions (centrum semiovale, basal ganglia, hippocampus, and midbrain) via a validated machine learning algorithm using T2-weighted MR images. Associations between sleep characteristics and perivascular space load were analysed with zero-inflated negative binomial regression models adjusted for various confounders. We found that higher actigraphy-estimated sleep efficiency was associated with a higher perivascular space load in the centrum semiovale (odds ratio 1.10, 95% confidence interval 1.04-1.16, p = 0.0008). No other actigraphic or polysomnographic sleep characteristics were associated with perivascular space load in other brain regions. We conclude that, contrary to our hypothesis, associations of sleep with perivascular space load in this middle-aged and elderly population remained limited to an association of a high actigraphy-estimated sleep efficiency with a higher perivascular space load in the centrum semiovale.
Collapse
Affiliation(s)
- Thom S Lysen
- Department of Epidemiology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Pinar Yilmaz
- Department of Epidemiology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Florian Dubost
- Biomedical Imaging Group Rotterdam, Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands.,Department of Neurology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Marleen de Bruijne
- Biomedical Imaging Group Rotterdam, Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands.,Department of Computer Science, University of Copenhagen, Copenhagen, Denmark
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Annemarie I Luik
- Department of Epidemiology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| |
Collapse
|
89
|
Novozhilova M, Mishchenko T, Kondakova E, Lavrova T, Gavrish M, Aferova S, Franceschi C, Vedunova M. Features of age-related response to sleep deprivation: in vivo experimental studies. Aging (Albany NY) 2021; 13:19108-19126. [PMID: 34320466 PMCID: PMC8386558 DOI: 10.18632/aging.203372] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/17/2021] [Indexed: 12/23/2022]
Abstract
Insomnia is currently considered one of the potential triggers of accelerated aging. The frequency of registered sleep-wake cycle complaints increases with age and correlates with the quality of life of elderly people. Nevertheless, whether insomnia is actually an age-associated process or whether it acts as an independent stress-factor that activates pathological processes, remains controversial. In this study, we analyzed the effects of long-term sleep deprivation modeling on the locomotor and orienting-exploratory activity, spatial learning abilities and working memory of C57BL/6 female mice of different ages. We also evaluated the modeled stress influence on morphological changes in brain tissue, the functional activity of the mitochondrial apparatus of nerve cells, and the level of DNA methylation and mRNA expression levels of the transcription factor HIF-1α (Hif1) and age-associated molecular marker PLIN2. Our findings point to the age-related adaptive capacity of female mice to the long-term sleep deprivation influence. For young (1.5 months) mice, the modeled sleep deprivation acts as a stress factor leading to weight loss against the background of increased food intake, the activation of animals' locomotor and exploratory activity, their mnestic functions, and molecular and cellular adaptive processes ensuring animal resistance both to stress and risk of accelerated aging development. Sleep deprivation in adult (7-9 months) mice is accompanied by an increase in body weight against the background of active food intake, increased locomotor and exploratory activity, gross disturbances in mnestic functions, and decreased adaptive capacity of brain cells, that potentially increasing the risk of pathological reactions and neurodegenerative processes.
Collapse
Affiliation(s)
- Maria Novozhilova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Tatiana Mishchenko
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Elena Kondakova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Tatiana Lavrova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Maria Gavrish
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Svetlana Aferova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Claudio Franceschi
- Institute of Information Technologies, Mathematics and Mechanics (ITMM), National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Maria Vedunova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| |
Collapse
|
90
|
Tuura RO, Volk C, Callaghan F, Jaramillo V, Huber R. Sleep-related and diurnal effects on brain diffusivity and cerebrospinal fluid flow. Neuroimage 2021; 241:118420. [PMID: 34302966 DOI: 10.1016/j.neuroimage.2021.118420] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/25/2021] [Accepted: 07/20/2021] [Indexed: 10/20/2022] Open
Abstract
The question of how waste products are cleared from the brain, and the role which sleep plays in this process, is critical for our understanding of a range of physical and mental illnesses. In rodents, both circadian and sleep-related processes appear to facilitate clearance of waste products. The purpose of this study was to investigate whether overnight changes in diffusivity, brain volumes, and cerebrospinal fluid flow measured with MRI are associated with sleep parameters from overnight high-density sleep EEG, and circadian markers. In healthy adults investigated with MRI before and after sleep EEG, we observed an increase in water diffusivity overnight, which was positively related to the proportion of total sleep time spent in rapid eye movement (REM) sleep, and negatively associated with the fraction of sleep time spent in non rapid eye movement (NREM) sleep. Diffusivity was also associated with the sleep midpoint, a circadian marker. CSF flow increased overnight; this increase was unrelated to sleep or diffusivity measures but was associated with circadian markers. These results provide evidence for both sleep related and diurnal effects on water compartmentalisation within the brain.
Collapse
Affiliation(s)
- Ruth O'Gorman Tuura
- Center for MR Research, University Children's Hospital, University of Zürich, Switzerland; Children's Research Center, University Children's Hospital, University of Zürich, Switzerland.
| | - Carina Volk
- Center for MR Research, University Children's Hospital, University of Zürich, Switzerland; Children's Research Center, University Children's Hospital, University of Zürich, Switzerland
| | - Fraser Callaghan
- Center for MR Research, University Children's Hospital, University of Zürich, Switzerland; Children's Research Center, University Children's Hospital, University of Zürich, Switzerland
| | - Valeria Jaramillo
- Children's Research Center, University Children's Hospital, University of Zürich, Switzerland; Child Development Center and Pediatric Sleep Center, University Children's Hospital, Zürich, Switzerland
| | - Reto Huber
- Children's Research Center, University Children's Hospital, University of Zürich, Switzerland; Child Development Center and Pediatric Sleep Center, University Children's Hospital, Zürich, Switzerland; Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| |
Collapse
|
91
|
Kopeikina E, Ponomarev ED. The Role of Platelets in the Stimulation of Neuronal Synaptic Plasticity, Electric Activity, and Oxidative Phosphorylation: Possibilities for New Therapy of Neurodegenerative Diseases. Front Cell Neurosci 2021; 15:680126. [PMID: 34335186 PMCID: PMC8318360 DOI: 10.3389/fncel.2021.680126] [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: 03/13/2021] [Accepted: 06/23/2021] [Indexed: 12/04/2022] Open
Abstract
The central nervous system (CNS) is highly vascularized where neuronal cells are located in proximity to endothelial cells, astroglial limitans, and neuronal processes constituting integrated neurovascular units. In contrast to many other organs, the CNS has a blood-brain barrier (BBB), which becomes compromised due to infection, neuroinflammation, neurodegeneration, traumatic brain injury, and other reasons. BBB disruption is presumably involved in neuronal injury during epilepsy and psychiatric disorders. Therefore, many types of neuropsychological disorders are accompanied by an increase in BBB permeability leading to direct contact of circulating blood cells in the capillaries with neuronal cells in the CNS. The second most abundant type of blood cells are platelets, which come after erythrocytes and outnumber ~100-fold circulating leukocytes. When BBB becomes compromised, platelets swiftly respond to the vascular injury and become engaged in thrombosis and hemostasis. However, more recent studies demonstrated that platelets could also enter CNS parenchyma and directly interact with neuronal cells. Within CNS, platelets become activated by recognizing major brain gangliosides on the surface of astrocytes and neurons and releasing a milieu of pro-inflammatory mediators, neurotrophic factors, and neurotransmitters. Platelet-derived factors directly stimulate neuronal electric and synaptic activity and promote the formation of new synapses and axonal regrowth near the site of damage. Despite such active involvement in response to CNS damage, the role of platelets in neurological disorders was not extensively studied, which will be the focus of this review.
Collapse
Affiliation(s)
| | - Eugene D. Ponomarev
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
92
|
The Serine Protease Homolog, Scarface, Is Sensitive to Nutrient Availability and Modulates the Development of the Drosophila Blood-Brain Barrier. J Neurosci 2021; 41:6430-6448. [PMID: 34210781 PMCID: PMC8318086 DOI: 10.1523/jneurosci.0452-20.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 02/08/2021] [Accepted: 03/14/2021] [Indexed: 01/21/2023] Open
Abstract
The adaptable transcriptional response to changes in food availability not only ensures animal survival but also lets embryonic development progress. Interestingly, the CNS is preferentially protected from periods of malnutrition, a phenomenon known as “brain sparing.” However, the mechanisms that mediate this response remain poorly understood. To get a better understanding of this, we used Drosophila melanogaster as a model, analyzing the transcriptional response of neural stem cells (neuroblasts) and glia of the blood–brain barrier (BBB) from larvae of both sexes during nutrient restriction using targeted DamID. We found differentially expressed genes in both neuroblasts and glia of the BBB, although the effect of nutrient deficiency was primarily observed in the BBB. We characterized the function of a nutritional sensitive gene expressed in the BBB, the serine protease homolog, scarface (scaf). Scaf is expressed in subperineurial glia in the BBB in response to nutrition. Tissue-specific knockdown of scaf increases subperineurial glia endoreplication and proliferation of perineurial glia in the blood–brain barrier. Furthermore, neuroblast proliferation is diminished on scaf knockdown in subperineurial glia. Interestingly, reexpression of Scaf in subperineurial glia is able to enhance neuroblast proliferation and brain growth of animals in starvation. Finally, we show that loss of scaf in the blood–brain barrier increases sensitivity to drugs in adulthood, suggesting a physiological impairment. We propose that Scaf integrates the nutrient status to modulate the balance between neurogenesis and growth of the BBB, preserving the proper equilibrium between the size of the barrier and the brain. SIGNIFICANCE STATEMENT The Drosophila BBB separates the CNS from the open circulatory system. The BBB glia are not only acting as a physical segregation of tissues but participate in the regulation of the metabolism and neurogenesis during development. Here we analyze the transcriptional response of the BBB glia to nutrient deprivation during larval development, a condition in which protective mechanisms are switched on in the brain. Our findings show that the gene scarface reduces growth in the BBB while promoting the proliferation of neural stem, assuring the balanced growth of the larval brain. Thus, Scarface would link animal nutrition with brain development, coordinating neurogenesis with the growth of the BBB.
Collapse
|
93
|
Semyachkina-Glushkovskaya O, Mamedova A, Vinnik V, Klimova M, Saranceva E, Ageev V, Yu T, Zhu D, Penzel T, Kurths J. Brain Mechanisms of COVID-19-Sleep Disorders. Int J Mol Sci 2021; 22:6917. [PMID: 34203143 PMCID: PMC8268116 DOI: 10.3390/ijms22136917] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
2020 and 2021 have been unprecedented years due to the rapid spread of the modified severe acute respiratory syndrome coronavirus around the world. The coronavirus disease 2019 (COVID-19) causes atypical infiltrated pneumonia with many neurological symptoms, and major sleep changes. The exposure of people to stress, such as social confinement and changes in daily routines, is accompanied by various sleep disturbances, known as 'coronasomnia' phenomenon. Sleep disorders induce neuroinflammation, which promotes the blood-brain barrier (BBB) disruption and entry of antigens and inflammatory factors into the brain. Here, we review findings and trends in sleep research in 2020-2021, demonstrating how COVID-19 and sleep disorders can induce BBB leakage via neuroinflammation, which might contribute to the 'coronasomnia' phenomenon. The new studies suggest that the control of sleep hygiene and quality should be incorporated into the rehabilitation of COVID-19 patients. We also discuss perspective strategies for the prevention of COVID-19-related BBB disorders. We demonstrate that sleep might be a novel biomarker of BBB leakage, and the analysis of sleep EEG patterns can be a breakthrough non-invasive technology for diagnosis of the COVID-19-caused BBB disruption.
Collapse
Affiliation(s)
- Oxana Semyachkina-Glushkovskaya
- Institute of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany;
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Aysel Mamedova
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Valeria Vinnik
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Maria Klimova
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Elena Saranceva
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Vasily Ageev
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Tingting Yu
- Wuhan National Laboratory for Optoelectronics, Britton Chance Center for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (T.Y.); (D.Z.)
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Dan Zhu
- Wuhan National Laboratory for Optoelectronics, Britton Chance Center for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (T.Y.); (D.Z.)
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Thomas Penzel
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
- Sleep Medicine Center, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jürgen Kurths
- Institute of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany;
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
- Potsdam Institute for Climate Impact Research, Telegrafenberg A31, 14473 Potsdam, Germany
| |
Collapse
|
94
|
Coll-Tané M, Gong NN, Belfer SJ, van Renssen LV, Kurtz-Nelson EC, Szuperak M, Eidhof I, van Reijmersdal B, Terwindt I, Durkin J, Verheij MMM, Kim CN, Hudac CM, Nowakowski TJ, Bernier RA, Pillen S, Earl RK, Eichler EE, Kleefstra T, Kayser MS, Schenck A. The CHD8/CHD7/Kismet family links blood-brain barrier glia and serotonin to ASD-associated sleep defects. SCIENCE ADVANCES 2021; 7:eabe2626. [PMID: 34088660 PMCID: PMC8177706 DOI: 10.1126/sciadv.abe2626] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 04/19/2021] [Indexed: 05/11/2023]
Abstract
Sleep disturbances in autism and neurodevelopmental disorders are common and adversely affect patient's quality of life, yet the underlying mechanisms are understudied. We found that individuals with mutations in CHD8, among the highest-confidence autism risk genes, or CHD7 suffer from disturbed sleep maintenance. These defects are recapitulated in Drosophila mutants affecting kismet, the sole CHD8/CHD7 ortholog. We show that Kismet is required in glia for early developmental and adult sleep architecture. This role localizes to subperineurial glia constituting the blood-brain barrier. We demonstrate that Kismet-related sleep disturbances are caused by high serotonin during development, paralleling a well-established but genetically unsolved autism endophenotype. Despite their developmental origin, Kismet's sleep architecture defects can be reversed in adulthood by a behavioral regime resembling human sleep restriction therapy. Our findings provide fundamental insights into glial regulation of sleep and propose a causal mechanistic link between the CHD8/CHD7/Kismet family, developmental hyperserotonemia, and autism-associated sleep disturbances.
Collapse
Affiliation(s)
- Mireia Coll-Tané
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, 6525 GA, Nijmegen, Netherlands.
| | - Naihua N Gong
- Departments of Psychiatry and Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Samuel J Belfer
- Departments of Psychiatry and Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lara V van Renssen
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, 6525 GA, Nijmegen, Netherlands
| | | | - Milan Szuperak
- Departments of Psychiatry and Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ilse Eidhof
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, 6525 GA, Nijmegen, Netherlands
| | - Boyd van Reijmersdal
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, 6525 GA, Nijmegen, Netherlands
| | - Isabel Terwindt
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, 6525 GA, Nijmegen, Netherlands
| | - Jaclyn Durkin
- Departments of Psychiatry and Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michel M M Verheij
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, Nijmegen, Netherlands
| | - Chang N Kim
- Departments of Anatomy and Psychiatry, University of California, San Francisco, CA 94143 USA
| | - Caitlin M Hudac
- Center for Youth Development and Intervention and Department of Psychology, University of Alabama, Tuscaloosa, AL 35487, USA
| | - Tomasz J Nowakowski
- Departments of Anatomy and Psychiatry, University of California, San Francisco, CA 94143 USA
| | - Raphael A Bernier
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98185, USA
| | - Sigrid Pillen
- Center for Sleep Medicine, Kempenhaeghe, Heeze, Netherlands
| | - Rachel K Earl
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98185, USA
| | - Evan E Eichler
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Tjitske Kleefstra
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, 6525 GA, Nijmegen, Netherlands
| | - Matthew S Kayser
- Departments of Psychiatry and Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, 6525 GA, Nijmegen, Netherlands.
| |
Collapse
|
95
|
Costa R. Frontiers in Chronobiology: Endogenous Clocks at the Core of Signaling Pathways in Physiology. Front Physiol 2021; 12:684745. [PMID: 34093241 PMCID: PMC8173170 DOI: 10.3389/fphys.2021.684745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/14/2021] [Indexed: 11/29/2022] Open
Affiliation(s)
- Rodolfo Costa
- Department of Biology, University of Padova, Italian National Research Council (CNR) Institute of Neuroscience, Padova, Italy
| |
Collapse
|
96
|
Naidu SAG, Wallace TC, Davies KJA, Naidu AS. Lactoferrin for Mental Health: Neuro-Redox Regulation and Neuroprotective Effects across the Blood-Brain Barrier with Special Reference to Neuro-COVID-19. J Diet Suppl 2021; 20:218-253. [PMID: 33977807 DOI: 10.1080/19390211.2021.1922567] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Overall mental health depends in part on the blood-brain barrier, which regulates nutrient transfer in-and-out of the brain and its central nervous system. Lactoferrin, an innate metal-transport protein, synthesized in the substantia nigra, particularly in dopaminergic neurons and activated microglia is vital for brain physiology. Lactoferrin rapidly crosses the blood-brain barrier via receptor-mediated transcytosis and accumulates in the brain capillary endothelial cells. Lactoferrin receptors are additionally present on glioma cells, brain micro-vessels, and neurons. As a regulator of neuro-redox, microglial lactoferrin is critical for protection/repair of neurons and healthy brain function. Iron imbalance and oxidative stress are common among patients with neurodegenerative disorders such as Parkinson's disease, Alzheimer's disease, dementia, depression, and multiple sclerosis. As an endogenous iron-chelator, lactoferrin prevents iron accumulation and dopamine depletion in Parkinson's disease patients. Oral lactoferrin supplementation could modulate the p-Akt/PTEN pathway, reduce Aβ deposition, and ameliorate cognitive decline in Alzheimer's disease. Novel lactoferrin-based nano-therapeutics have emerged as effective drug-delivery systems for clinical management of neurodegenerative disorders. Recent emergence of the Coronavirus disease-2019 (COVID-19) pandemic, initially considered a respiratory illness, demonstrated a broader virulence spectrum with the ability to cross the blood-brain barrier and inflict a plethora of neuropathological manifestations in the brain - the Neuro-COVID-19. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections are widely reported in Parkinson's disease, Alzheimer's disease, dementia, and multiple sclerosis patients with aggravated clinical outcomes. Lactoferrin, credited with several neuroprotective benefits in the brain could serve as a potential adjuvant in the clinical management of Neuro-COVID-19.
Collapse
Affiliation(s)
- Sreus A G Naidu
- N-terminus Research Laboratory, Yorba Linda, California, USA
| | - Taylor C Wallace
- Department of Nutrition and Food Studies, George Mason University, Fairfax, Virginia, USA
- Think Healthy Group, Washington, District of Columbia, USA
| | - Kelvin J A Davies
- Division of Biogerontology, Leonard Davis School of Gerontology, The University of Southern California, Los Angeles, California, USA
- Division of Molecular & Computational Biology, Dornsife College of Letters, Arts, and Sciences, The University of Southern California, Los Angeles, California, USA
- Department Biochemistry & Molecular Medicine, Keck School of Medicine of USC, The University of Southern California, Los Angeles, California, USA
| | | |
Collapse
|
97
|
Lo EH, Albers GW, Dichgans M, Donnan G, Esposito E, Foster R, Howells DW, Huang YG, Ji X, Klerman EB, Lee S, Li W, Liebeskind DS, Lizasoain I, Mandeville ET, Moro MA, Ning M, Ray D, Sakadžić S, Saver JL, Scheer FAJL, Selim M, Tiedt S, Zhang F, Buchan AM. Circadian Biology and Stroke. Stroke 2021; 52:2180-2190. [PMID: 33940951 DOI: 10.1161/strokeaha.120.031742] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Circadian biology modulates almost all aspects of mammalian physiology, disease, and response to therapies. Emerging data suggest that circadian biology may significantly affect the mechanisms of susceptibility, injury, recovery, and the response to therapy in stroke. In this review/perspective, we survey the accumulating literature and attempt to connect molecular, cellular, and physiological pathways in circadian biology to clinical consequences in stroke. Accounting for the complex and multifactorial effects of circadian rhythm may improve translational opportunities for stroke diagnostics and therapeutics.
Collapse
Affiliation(s)
- Eng H Lo
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Departments of Radiology (E.H.L., E.E., W.L., E.T.M., S.S., F.Z.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Gregory W Albers
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Department of Neurology, Stanford Stroke Center, Stanford University, Palo Alto (G.W.A., S.L.)
| | - Martin Dichgans
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,German Center for Neurodegenerative Diseases (DZNE, Munich) and Munich Cluster for Systems Neurology (SyNergy), Germany (M.D.).,Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Germany (M.D., S.T.)
| | - Geoffrey Donnan
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Departments of Medicine and Neurology, Royal Melbourne Hospital, University of Melbourne, Australia (G.D.)
| | - Elga Esposito
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Departments of Radiology (E.H.L., E.E., W.L., E.T.M., S.S., F.Z.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Russell Foster
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences (R.F.), University of Oxford, United Kingdom
| | - David W Howells
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Tasmanian School of Medicine, University of Tasmania, Australia (D.W.H.)
| | - Yi-Ge Huang
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Department of Stroke Medicine (Y.H., A.M.B.), University of Oxford, United Kingdom
| | - Xunming Ji
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Beijing Institute for Brain Disorders, China (X.J.)
| | - Elizabeth B Klerman
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Neurology (E.B.K., M.N.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Sarah Lee
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Department of Neurology, Stanford Stroke Center, Stanford University, Palo Alto (G.W.A., S.L.)
| | - Wenlu Li
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Departments of Radiology (E.H.L., E.E., W.L., E.T.M., S.S., F.Z.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - David S Liebeskind
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Department of Neurology, Geffen School of Medicine, University of California Los Angeles (J.L.S., D.S.L.)
| | - Ignacio Lizasoain
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Department of Pharmacology and Toxicology, Complutense Medical School, Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain (I.L.)
| | - Emiri T Mandeville
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Departments of Radiology (E.H.L., E.E., W.L., E.T.M., S.S., F.Z.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Maria A Moro
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid, Spain (M.A.M.)
| | - MingMing Ning
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Neurology (E.B.K., M.N.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - David Ray
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, and Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, United Kingdom (D.R.)
| | - Sava Sakadžić
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Departments of Radiology (E.H.L., E.E., W.L., E.T.M., S.S., F.Z.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Jeffrey L Saver
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Department of Neurology, Geffen School of Medicine, University of California Los Angeles (J.L.S., D.S.L.)
| | - Frank A J L Scheer
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Departments of Medicine and Neurology, Brigham & Women's Hospital (F.A.J.L.S.), Harvard Medical School, Boston
| | - Magdy Selim
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Department of Neurology, Beth Israel Deaconess Medical Center (M.S.), Harvard Medical School, Boston
| | - Steffen Tiedt
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Germany (M.D., S.T.)
| | - Fang Zhang
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Departments of Radiology (E.H.L., E.E., W.L., E.T.M., S.S., F.Z.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Alastair M Buchan
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Department of Stroke Medicine (Y.H., A.M.B.), University of Oxford, United Kingdom
| |
Collapse
|
98
|
Lee S, Jung W, Eom S, Yeom HD, Park HD, Lee JH. Molecular Regulation of Betulinic Acid on α3β4 Nicotinic Acetylcholine Receptors. Molecules 2021; 26:2659. [PMID: 34062829 PMCID: PMC8125762 DOI: 10.3390/molecules26092659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/17/2021] [Accepted: 04/26/2021] [Indexed: 11/24/2022] Open
Abstract
Betulinic acid (BA) is a major constituent of Zizyphus seeds that have been long used as therapeutic agents for sleep-related issues in Asia. BA is a pentacyclic triterpenoid. It also possesses various anti-cancer and anti-inflammatory effects. Current commercially available sleep aids typically use GABAergic regulation, for which many studies are being actively conducted. However, few studies have focused on acetylcholine receptors that regulate wakefulness. In this study, we utilized BA as an antagonist of α3β4 nicotinic acetylcholine receptors (α3β4 nAChRs) known to regulate rapid-eye-movement (REM) sleep and wakefulness. Effects of BA on α3β4 nAChRs were concentration-dependent, reversible, voltage-independent, and non-competitive. Site-directed mutagenesis and molecular-docking studies confirmed the binding of BA at the molecular level and showed that the α3 subunit L257 and the β4 subunit I263 residues affected BA binding. These data demonstrate that BA can bind to a binding site different from the site for the receptor's ligand, acetylcholine (ACh). This suggests that BA may be an effective antagonist that is unaffected by large amounts of ACh released during wakefulness and REM sleep. Based on the above experimental results, BA is likely to be a therapeutically useful sleep aid and sedative.
Collapse
Affiliation(s)
- Shinhui Lee
- Department of Biotechnology, Chonnam National University, Gwangju 61186, Korea; (S.L.); (S.E.)
| | - Woog Jung
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Korea;
| | - Sanung Eom
- Department of Biotechnology, Chonnam National University, Gwangju 61186, Korea; (S.L.); (S.E.)
| | | | - Heui-Dong Park
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Korea;
| | - Junho H. Lee
- Department of Biotechnology, Chonnam National University, Gwangju 61186, Korea; (S.L.); (S.E.)
| |
Collapse
|
99
|
Björk V. Aging of the Suprachiasmatic Nucleus, CIRCLONSA Syndrome, Implications for Regenerative Medicine and Restoration of the Master Body Clock. Rejuvenation Res 2021; 24:274-282. [PMID: 33573456 DOI: 10.1089/rej.2020.2388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The suprachiasmatic nucleus (SCN) in the brain is the master regulator of the circadian clocks throughout the human body. With increasing age the circadian clock in humans and other mammals becomes increasingly disorganized leading to a large number of more or less well-categorized problems. While a lot of aging research has focused on the peripheral clocks in tissues across organisms, it remains a paramount task to quantify aging of the most important master clock, the human SCN. Furthermore, a pipeline needs to be developed with therapies to mitigate the systemic cellular circadian dysfunction in the elderly and ultimately repair and reverse aging of the SCN itself. A disease classification for the aging SCN, Circadian Clock Neuronal Senile Atrophy (CIRCLONSA syndrome), would improve research funding and goal-oriented biotechnological entrepreneurship.
Collapse
|
100
|
Rigon L, De Filippis C, Napoli B, Tomanin R, Orso G. Exploiting the Potential of Drosophila Models in Lysosomal Storage Disorders: Pathological Mechanisms and Drug Discovery. Biomedicines 2021; 9:biomedicines9030268. [PMID: 33800050 PMCID: PMC8000850 DOI: 10.3390/biomedicines9030268] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/18/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
Lysosomal storage disorders (LSDs) represent a complex and heterogeneous group of rare genetic diseases due to mutations in genes coding for lysosomal enzymes, membrane proteins or transporters. This leads to the accumulation of undegraded materials within lysosomes and a broad range of severe clinical features, often including the impairment of central nervous system (CNS). When available, enzyme replacement therapy slows the disease progression although it is not curative; also, most recombinant enzymes cannot cross the blood-brain barrier, leaving the CNS untreated. The inefficient degradative capability of the lysosomes has a negative impact on the flux through the endolysosomal and autophagic pathways; therefore, dysregulation of these pathways is increasingly emerging as a relevant disease mechanism in LSDs. In the last twenty years, different LSD Drosophila models have been generated, mainly for diseases presenting with neurological involvement. The fruit fly provides a large selection of tools to investigate lysosomes, autophagy and endocytic pathways in vivo, as well as to analyse neuronal and glial cells. The possibility to use Drosophila in drug repurposing and discovery makes it an attractive model for LSDs lacking effective therapies. Here, ee describe the major cellular pathways implicated in LSDs pathogenesis, the approaches available for their study and the Drosophila models developed for these diseases. Finally, we highlight a possible use of LSDs Drosophila models for drug screening studies.
Collapse
Affiliation(s)
- Laura Rigon
- Fondazione Istituto di Ricerca Pediatrica “Città della Speranza”, Corso Stati Uniti 4, 35127 Padova, Italy; (C.D.F.); (R.T.)
- Correspondence:
| | - Concetta De Filippis
- Fondazione Istituto di Ricerca Pediatrica “Città della Speranza”, Corso Stati Uniti 4, 35127 Padova, Italy; (C.D.F.); (R.T.)
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Department of Women’s and Children’s Health, University of Padova, Via Giustiniani 3, 35128 Padova, Italy
| | - Barbara Napoli
- Laboratory of Molecular Biology, Scientific Institute, IRCCS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, 23842 Lecco, Italy;
| | - Rosella Tomanin
- Fondazione Istituto di Ricerca Pediatrica “Città della Speranza”, Corso Stati Uniti 4, 35127 Padova, Italy; (C.D.F.); (R.T.)
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Department of Women’s and Children’s Health, University of Padova, Via Giustiniani 3, 35128 Padova, Italy
| | - Genny Orso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy;
| |
Collapse
|